WO1999024029A1 - Process for producing arsenic trioxide formulations and methods for treating cancer using arsenic trioxide or melarsoprol - Google Patents

Process for producing arsenic trioxide formulations and methods for treating cancer using arsenic trioxide or melarsoprol Download PDF

Info

Publication number
WO1999024029A1
WO1999024029A1 PCT/US1998/024024 US9824024W WO9924029A1 WO 1999024029 A1 WO1999024029 A1 WO 1999024029A1 US 9824024 W US9824024 W US 9824024W WO 9924029 A1 WO9924029 A1 WO 9924029A1
Authority
WO
WIPO (PCT)
Prior art keywords
arsenic trioxide
human
arsenic
melarsoprol
leukemia
Prior art date
Application number
PCT/US1998/024024
Other languages
French (fr)
Inventor
Raymond P. Warrell, Jr.
Pier Paolo Pandolfi
Janice L. Gabrilove
Original Assignee
Memorial Sloan-Kettering Cancer Center
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=22057410&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO1999024029(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to NZ504585A priority Critical patent/NZ504585A/en
Priority to AU13973/99A priority patent/AU747474C/en
Priority to KR1020007005045A priority patent/KR20010031938A/en
Priority to JP2000520121A priority patent/JP2001522799A/en
Priority to PL98343868A priority patent/PL343868A1/en
Application filed by Memorial Sloan-Kettering Cancer Center filed Critical Memorial Sloan-Kettering Cancer Center
Priority to EP98957803A priority patent/EP1037625A4/en
Priority to BR9814857-5A priority patent/BR9814857A/en
Priority to IL13605198A priority patent/IL136051A0/en
Priority to CA2309652A priority patent/CA2309652C/en
Publication of WO1999024029A1 publication Critical patent/WO1999024029A1/en
Priority to NO20002409A priority patent/NO20002409L/en
Priority to AU2002300339A priority patent/AU2002300339B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/53Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with three nitrogens as the only ring hetero atoms, e.g. chlorazanil, melamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/28Compounds containing heavy metals
    • A61K31/285Arsenic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/555Heterocyclic compounds containing heavy metals, e.g. hemin, hematin, melarsoprol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/36Arsenic; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K41/00Medicinal preparations obtained by treating materials with wave energy or particle radiation ; Therapies using these preparations
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention relates to methods and compositions for the treatment of leukemia, lymphoma, and certain other cancers.
  • the present invention relates to the novel uses of arsenic trioxide and an organic arsenic compound for treating acute leukemia and chronic leukemia.
  • Cancer is characterized primarily by an increase in the number of abnormal cells derived from a given normal tissue, invasion of adjacent tissues by these abnormal cells, and lymphatic or blood-borne spread of malignant cells to regional lymph nodes and to distant sites (metastasis) .
  • Clinical data and molecular biologic studies indicate that cancer is a multistep process that begins with minor preneoplastic changes, which may under certain conditions progress to neoplasia.
  • neoplastic lesion may evolve clonally to grow into a solid tumor, and develop an increasing capacity for invasion, growth, metastasis, and heterogeneity, especially under conditions in which the neoplastic cells escape the host's immune surveillance (Roitt, I., Brostoff, J and Kale, D. , 1993, Immunology, 3rd ed.
  • Leukemia refers to malignant neoplasms of the blood-forming tissues. Transformation to malignancy typically occurs in a single cell through two or more steps with subsequent proliferation and clonal expansion. In some leukemias, specific chromosomal translocations have been identified with consistent leukemic cell morphology and special clinical features (e.g., translocations of 9 and 22 in chronic myelocytic leukemia, and of 15 and 17 in acute promyelocytic leukemia) . Acute leukemias are predominantly undifferentiated cell populations and chronic leukemias more mature cell forms.
  • Acute leukemias are divided into lymphoblastic (ALL) and non-lymphoblastic (ANLL) types. They may be further subdivided by their morphologic and cytochemical appearance according to the French-American-British (FAB) classification or according to their type and degree of differentiation. The use of specific B- and T-cell and myeloid-antigen monoclonal antibodies are most helpful for classification.
  • ALL is predominantly a childhood disease which is established by laboratory findings and bone marrow examination.
  • ANLL also known as acute myeloblastic leukemia (AML) , occurs at all ages and is the more common acute leukemia among adults; it is the form usually associated with irradiation as a causative agent.
  • AML acute myeloblastic leukemia
  • CLL lymphocytic
  • CML myelocytic
  • CLL is characterized by the appearance of mature lymphocytes in blood, bone marrow, and lymphoid organs. The hallmark of CLL is sustained, absolute lymphocytosis (> 5,000/ ⁇ L) and an increase of lymphocytes in the bone marrow. Most CLL patients also have clonal expansion of lymphocytes with B- cell characteristics.
  • CLL is a disease of older persons.
  • the characteristic feature is the predominance of granulocytic cells of all stages of differentiation in blood, bone marrow, liver, spleen, and other organs. In the symptomatic patient at diagnosis the total BC count is usually about 200,000/ ⁇ L, but may reach 1, 000, 000/ ⁇ L.
  • CML is relatively easy to diagnose because of the presence of the Philadelphia chromosome.
  • hematopoietic cancer necessitates using systemic chemotherapy as the primary treatment modality.
  • Drugs selected according to sensitivities of specific leukemias are usually given in combination.
  • Radiation therapy may be used as an adjunct to treat local accumulations of leukemic cells.
  • Surgery is rarely indicated as a primary treatment modality, but may be used in managing some complications. Bone marrow transplantation from an HLA-matched sibling is sometimes indicated.
  • Arsenic has been considered to be both a poison and a drug for a long time in both Western and Chinese medical practices. In the latter part of the nineteenth century, arsenic was used frequently in attempts to treat diseases of the blood in the West. In 1878, it was reported that treatment of a leukemic patient with Fowler's solution (a solution containing potassium arsenite, valence +5) reduced markedly the count of white blood cells (Cutler and Bradford, Am. J. Med. Sci.. January 1878, 81-84).
  • Fowler's solution a solution containing potassium arsenite, valence +5
  • Fowler's solution as a palliative agent to treat chronic myelogenous leukemia (CML) was described by Forkner and Scott in 1931 (J. Am. Med. Assoc. , 1931, iii, 97) , and later confirmed by Stephens and Lawrence in 1936 (Ann. Intern. Med. 9, 1488-1502).
  • active chemical ingredient (s) of Fowler's solution was not determined, its toxicity was well recognized.
  • Fowler's solution was administered strictly as an oral composition, and was given to leukemic patients as a solution until the level of white blood cells was depressed to an acceptable level or until toxicities (such as skin keratoses and hyperpigmentation) developed, while the patients enjoyed varying periods of remission.
  • toxicities such as skin keratoses and hyperpigmentation
  • Fowler's solution was still used occasionally in attempts to treat CML, however, most patients with CML were treated with other chemotherapeutic agents, such as busulfan, and/or radiation therapy (Monfardini et al . , Cancer. 1973, 31:492-501).
  • arsenous acid or arsenic trioxide paste has been used to treat tooth marrow diseases, psoriasis, syphilis and rheumatosis (Chen et al . ,
  • arsenic trioxide had been applied experimentally to treat acute promyelocytic leukemia (APL) in China (commented by Mervis, 1996, Science. 273:578).
  • APL acute promyelocytic leukemia
  • the clinical efficacy of arsenic trioxide has recently been re-investigated in 14 of 15 patients with refractory APL, where the use of an intravenous dose at 10 mg/day for 4-9 weeks was reported to result in complete morphologic remission without associated bone marrow suppression (Shen et al . , 1997, Blood, 89:3354-
  • arsenic is due to the ability of arsenic to direct the nucleoplasmic fraction of PML to nuclear bodies for degradation (Zhu et al., 1997, Proc. Natl. Acad. Sci., 94:3978-3983).
  • arsenic is well known to be both a poison and a carcinogenic agent, there have been many reports concerning the use of arsenic in medical treatment.
  • leukemias there are many different types of leukemias, each of which requires a unique treatment protocol that is modified according to the presence of factors predicting for a risk of treatment failure.
  • the development of a broad spectrum anti- leukemia agent that can be used alone or in combination with other existing drugs is extremely desirable.
  • arsenic rioxide and the organic arsenical, melarsoprol have broad applicability in the treatment of various types of leukemias, lymphomas, and solid tumors.
  • the invention described herein encompasses a method of treating leukemia, lymphoma or solid tumors comprising the administration of a therapeutically effective and non-lethal amount of arsenic trioxide or melarsoprol to a human in need of such therapy.
  • the invention also encompasses the use of combination therapy to treat leukemia, especially leukemias which are refractory to other forms of treatment.
  • the invention also encompasses a method for the manufacture of pharmaceutical compositions comprising arsenic trioxide.
  • arsenic trioxide or melarsoprol compounds can be used alone or in combination with other known therapeutic agents (including chemotherapeutics, radioprotectants and radiotherapeutics) or techniques to either improve the quality of life of the patient, or to treat leukemia, lymphoma or solid tumor.
  • the arsenic compounds can be used before, during or after the administration of one or more known chemotherapeutic agents, including antitumor agents.
  • the arsenic compounds can be used before, during or after radiation treatment.
  • the pharmaceutical compositions of the invention are sterile solutions suitable for intravenous injection or infusion.
  • the invention encompasses a composition suitable for oral delivery; comprising arsenic trioxide or melarsoprol and a pharmaceutically acceptable excipient or carrier.
  • the invention also includes compositions suitable for topical or transdermal delivery, including but not limited to iontophoretic methods. Specific therapeutic regimens, pharmaceutical compositions, and kits are also provided by the invention.
  • This invention provides a method of treating acute or chronic leukemia, lymphoma, or solid tumors in a human which comprises administering to a human in need of such therapy a therapeutically effective and non-lethal amount of one or more arsenic compounds, such as arsenic trioxide or melarsoprol.
  • the invention also includes a method of treating leukemia in a human who has become refractory to other forms of treatment which comprises administering to a human arsenic trioxide or melarsoprol in combination with another chemotherapeutic agent, e.g., all-trans retinoic acid (ATRA) .
  • ATRA all-trans retinoic acid
  • the invention also relates to a method for the manufacture of pharmaceutical compositions comprising arsenic trioxide. It is preferred that pharmaceutical compositions of the present invention exhibit reduced toxicity, improved efficacy, improved stability during storage and use, and that the composition has a physiologically acceptable pH.
  • aromatic compound refers to a pharmaceutically acceptable form of arsenic trioxide (As0 3 ) or melarsoprol.
  • Melarsoprol is an organic arsenic compound which can be synthesized by complexing melarsen oxide with dimercaprol or commercially purchased (Arsobal® by Rhone Poulenc Rorer, Collegeville, PA) . Since the non- pharmaceutically formulated raw materials of the invention are well known, they can be prepared from well-known chemical techniques in the art. (See for example , Kirk-Othmer,
  • chemotherapeutic regimen means conventional drugs and drug therapies, including vaccines, for treating cancer, viral infections, and other malignancies, which are known to those skilled in the art.
  • radiotherapeutic agents are well known in the art.
  • arsenic trioxide or melarsoprol compounds can be used alone or in combination with other known therapeutic agents (including chemotherapeutics, radioprotectants and radiotherapeutics) or techniques to either improve the quality of life of the patient, or to treat leukemia, lymphoma or solid tumor.
  • the arsenic compounds can be used before, during or after the administration of one or more known antitumor agents including but not limited to mustard compounds, nitrogen mustard, chlorambucil, melphalan, cyclophosphamide, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil, floxuridine, methotrexate, vincristine, vinblastine, taxol, etoposide, temiposide, dactinomycin, daunorubicin, doxorubicin, bleomycin, mitomycin, cisplatin, carboplatin, estramustine phosphate, hydroxyurea, BCNU, procarbazine, VM- 26, interferons, and all-trans retinoic acid (ATRA) , or other retinoids (See, for example , the Physician Desk References 1997) .
  • the arsenic compounds can be used before, during or after radiation treatment.
  • the arsenic compound of the invention and ATRA can be administered as a mixture.
  • the lymphoma, leukemia or solid tumor in the human treated by the combination is refractory to general methods of treatment, or is a relapsed case of leukemia.
  • any suitable mode of administration may be used in accordance with the present invention including but not limited to parenteral administration such as intravenous, subcutaneous, intramuscular and intrathecal administration; oral, and intranasal administration, and inhalation.
  • parenteral administration such as intravenous, subcutaneous, intramuscular and intrathecal administration; oral, and intranasal administration, and inhalation.
  • the mode of administration will vary according to the type of cancer, and the condition of the human.
  • compositions to be used may be in the form of sterile aqueous or organic solutions, colloidal suspensions, caplets, tablets and cachets.
  • a method for treating leukemia means that the disease and the symptoms associated with the disease are alleviated, reduced, cured, or placed in a state of remission.
  • the methods of treatment of the invention can lower the white blood cell count, or reduce lymphocytosis in a human under treatment.
  • a method for treating lymphoma means that the disease and the symptoms associated with the disease are alleviated, reduced, cured, or placed in a state of remission.
  • a method for treating solid tumor means that the disease and the symptoms associated with the solid tumor are alleviated, reduced, cured, or placed in a state of remission.
  • a method for treating leukemic infiltration means that the infiltration of leukemic cells out of circulation and into other organs and systems and the symptoms associated with such infiltration are alleviated, reduced, cured, or placed in a state of remission.
  • refractory when used herein means that the leukemia is generally resistant to treatment or cure.
  • preneoplastic cell refers to a cell which is in transition from a normal to a neoplastic form; or cells that fail to differentiate normally; and morphological evidence, increasingly supported by molecular biologic studies, indicates that preneoplasia progresses through multiple steps.
  • the invention provides a method for treatment of leukemia in a human comprising the administration of a therapeutically effective and non-lethal amount of arsenic trioxide or melarsoprol to the human.
  • the invention also provides a weight-based dosing regimen, not heretofore disclosed, that maximizes the safety in humans of these otherwise highly toxic compounds.
  • Arsenic trioxide inhibits growth and induce apoptosis in NB4 acute promyelocytic leukemic cells.
  • Acute promyelocytic leukemia (APL) is associated with the t(15;17) translocation, which generates a PML/RAR fusion protein between PML, a growth suppressor localized on nuclear matrix- associated bodies, and RAR ⁇ , a nuclear receptor for retinoic acid (RA) .
  • PML/RAR ⁇ was proposed to block myeloid differentiation through inhibition of nuclear receptor response, as does a dominant negative RAR ⁇ mutant.
  • PML/RAR ⁇ displaces PML and other nuclear body (NB) antigens onto nuclear microspeckles, likely resulting in the loss of PML and/or NB functions. It has been suggested that high concentrations of arsenic trioxide promote apoptosis, whereas low concentrations induce partial differentiation in NB4 cells as well as cells derived from APL patients. It was postulated that As 2 0 3 works through its ability to specifically cause PML-RAR ⁇ in APL cells to be relocalized to nuclear bodies for degradation (Zhu et al. , 1997, Proc. Natl. Acad. Sci . USA. 94:3978-3983).
  • arsenic compounds of the present invention can be used against a variety of leukemias, including but not limited to:
  • ALL Acute lymphoblastic leukemia
  • AML Acute yeloblastic leukemia
  • APL Acute promyelocytic leukemia
  • Acute monoblastic leukemia Acute erythroleukemic leukemia
  • CML Chronic myelocytic leukemia
  • CLL Chronic lymphocytic leukemia
  • the invention provides a method for treatment of lymphoma in a human comprising the administration of a therapeutically effective and non-lethal amount of arsenic trioxide or melarsoprol to the human.
  • Lymphoma that can be treated by the methods of the invention include but are not limited to high grade lymphoma, intermediate grade lymphoma, low grade lymphoma, and the various subclassifications.
  • the invention provides a method for treatment of solid tumors, including metastasises, in humans comprising the administration of a therapeutically effective and non-lethal amount of arsenic trioxide or melarsoprol to the human.
  • Solid tumors that can be treated by the methods of the invention include but are not limited to: cancer of the digestive tract, oesophagus, liver, stomach, and colon; skin; brain; bone; breast; lung; and soft tissues, including but not limited to various sarcomas, and preferably prostate cancer.
  • the leukemic or tumor cells are infiltrating other organs and systems in a human, for example, the central nervous system.
  • the methods of the invention are also applicable to reduce the number of preneoplastic cells in a human in which there is an abnormal increase in the number of preneoplastic cells.
  • the invention provides a method of treatment of acute promyelolytic leukemia (APL) in a human comprising the administration of a therapeutically effective and non-lethal amount of melarsoprol to the human.
  • APL acute promyelolytic leukemia
  • the inventors discovered, as described in Section 5.2, that concentrations of melarsoprol that are cytotoxic in vitro can readily be achieved in vivo .
  • the invention provides a method of treatment of chronic myelogenous leukemia (CML) in a human comprising the administration of a therapeutically effective and non-lethal amount of arsenic trioxide to the human.
  • CML chronic myelogenous leukemia
  • the inventors discovered, as described in Section 5.3, that arsenic trioxide can also induce apoptosis in a CML cell line.
  • the therapeutic benefits of the pharmaceutical compositions of the invention comprising arsenic trioxide is far superior to that of potassium arsenite, commonly formulated as Fowler's solution.
  • the invention provides a method of treatment of acute promyelocytic leukemia (APL) in a human, in which the APL is associated with a translocation of the RAR ⁇ locus on chromosome 17 to chromosome 11, comprising the administration of a therapeutically effective amount of arsenic trioxide or melarsoprol to the human.
  • APL acute promyelocytic leukemia
  • RAR ⁇ on chromosome 17 translocates and fuses with the PML gene located on chromosome 15, i.e., t(15;l7).
  • RAR ⁇ translocates to chromosome 11 where it fuses to the PLZF gene.
  • APL associated with the t(ll;17) shows a distinctly worse prognosis with poor response to chemotherapy and little or no response to treatment with ATRA, thus defining a new APL syndrome.
  • the present invention provides that arsenic trioxide or melarsoprol can be used to treat such cases of APL.
  • Transgenic animal models of APL associated with t(15;17) and t(ll;17) for testing the therapeutic benefits and dosages of arsenic compounds of the invention are described in Section 5.4 hereinbelow.
  • the invention provides a method of treatment of leukemia in a human who is not responding to conventional therapy comprising the administration of a therapeutically effective and non-lethal amount of a combination of arsenic compound and another chemotherapeutic agent, such as but not limited to, all-trans retinoic acid (ATRA) or other retinoids, to the human.
  • ATRA all-trans retinoic acid
  • the arsenic compound can either be arsenic trioxide or melarsoprol or a pharmaceutically acceptable salt thereof.
  • the invention also encompasses the treatment of retinoid- resistant patients with an arsenic compound.
  • the arsenic compound of the invention and the chemotherapeutic agent can be administered either as a mixture or sequentially.
  • the arsenic compound may be administered before or after the chemotherapeutic agent, so long as the first administered agent is still providing antileukemic activity in the human when the second agent is administered.
  • Any of the modes of administration described herein may be used to deliver the combination.
  • the leukemia in the human treated by the combination is refractory to general methods of treatment, or is a relapsed case of leukemia.
  • compositions comprising a compound of the invention formulated in a compatible pharmaceutical carrier may be prepared, packaged, labelled for treatment of and used for the treatment of the indicated leukemia, lymphoma, or solid tumor.
  • the invention provides a method for the manufacture of a pharmaceutical composition comprising a therapeutic effective and non-lethal amount of arsenic trioxide (As : 0 3 ) .
  • Arsenic trioxide raw material
  • Arsenic trioxide is a solid inorganic compound that is commercially available in a very pure form. However, it is difficult to dissolve As : 0, in aqueous solution. Further, the inventors are unaware of any published teachings on how to formulate As : 0, as a pharmaceutical composition suitable for injection directly into a human.
  • Arsenic is present in solution in the +5 valence state (pentavalent) or the +3 valence state (trivalent) .
  • potassium arsenite (KAsO, ; which is present in Fowler's solution) and salts of arsenious acid contain pentavalent arsenic. It is known that one form of arsenic is more toxic than the other. (Goodman & Gilman's The Pharmacological Basis of Therapeutics, 9th edition, chapter 66, 1660, 1997) .
  • a fresh solution of arsenic trioxide containing arsenic in the trivalent state will be gradually oxidized to pentavalent state if exposed to air for a prolonged period, and as a result of the accumulation of pentavalent arsenic, the relative toxicity of a solution of As : 0 3 will change over time. (Id .
  • the inventors have experimented and successfully developed a method for formulating arsenic trioxide which overcomes the above-described problems of solubility and stability.
  • the method comprises solubilizing solid high purity As : 0 ? in an aqueous solution at high pH, such as pH greater than 12.
  • a 5 M solution of sodium hydroxide may be used.
  • mechanical stirring and/or gentle heating may be applied.
  • a solution of As 2 0. can also be obtained by dissolving the solid compound overnight.
  • a solution of 1 M As : 0. is obtained by this method.
  • this solution is too basic to be useful as a pharmaceutical composition.
  • the solution is first diluted in water, for example, to a concentration of about 1 mg/mL, pH 12.
  • the As : 0. solution is then back- titrated with acid, such as, hydrochloric acid (1 M to 5 M HC1) , with constant stirring until the pH is about 8.0 to 8.5.
  • acid such as, hydrochloric acid (1 M to 5 M HC1)
  • Highly concentrated HCl is not suitab.le as it causes precipitation to occur in the As0 ; . solution.
  • the partially neutralized As-0 ? solution may then be sterilized for example, by filtration (e.g., through a 0.22 ⁇ m filter), and stored in sterile vials.
  • composition that can be directly injected into a subject, the composition must be sterile, standard techniques known to the skilled artisan for sterilization can be used. See, e.g., Remington's
  • the pH of the partially neutralized As : 0. solution may be further adjusted to near physiological pH by dilution (10-100 fold) with a pharmaceutical carrier, such as a 5% dextrose solution.
  • a pharmaceutical carrier such as a 5% dextrose solution.
  • lOmL of a partially neutralized As : 0 solution can be added to 500 mL of a 5% dextrose solution to yield a final pH of about 6.5 to 7.5.
  • the method of the invention reduces the oxidation of arsenic in solution.
  • Pharmaceutical compositions containing arsenic trioxide manufactured by the method of the invention show improved stability and long shelf life.
  • the arsenic compounds and their physiologically acceptable solvates may be formulated for oral or parenteral administration.
  • the pharmaceutical preparation may be in liquid form, for example, solutions, syrups or suspensions, or may be presented as a drug product for reconstitution with water or other suitable vehicle before use.
  • Such liquid preparations may be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e.g., sorbitol syrup, cellulose derivatives or hydrogenated edible fats) ; emulsifying agents (e .g . , lecithin or acacia); non-aqueous vehicles (e . g. , almond oil, oily esters, or fractionated vegetable oils) ; and preservatives (e . g . , methyl or propyl-p-hydroxybenzoates or sorbic acid) .
  • suspending agents e.g., sorbitol syrup, cellulose derivatives or hydrogenated edible fats
  • emulsifying agents e .g . , lecithin or acacia
  • non-aqueous vehicles e g. , almond oil,
  • the pharmaceutical compositions may take the form of, for example, tablets or capsules prepared by conventional means with pharmaceutically acceptable excipients such as binding agents (e . g . , pregelatinized maize starch, polyvinyl pyrrolidone or hydroxypropyl methylcellulose) ; fillers (e.g., lactose, microcrystalline cellulose or calcium hydrogen phosphate); lubricants (e.g., magnesium stearate, talc or silica); disintegrants (e.g., potato starch or sodium starch glycolate) ; or wetting agents (e.g., sodium lauryl sulphate).
  • binding agents e . g . , pregelatinized maize starch, polyvinyl pyrrolidone or hydroxypropyl methylcellulose
  • fillers e.g., lactose, microcrystalline cellulose or calcium hydrogen phosphate
  • lubricants e.g., magnesium stearate, talc or silica
  • the compounds for use according to the present invention are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • Capsules and cartridges of, e .g. , gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
  • the compounds may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion.
  • Formulations for injection may be presented in unit dosage form, e.g., in ampules or in multi- dose containers, with an added preservative.
  • the compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • the invention also provides kits for carrying out the therapeutic regimens of the invention.
  • kits comprise in one or more containers therapeutically effective amounts of the arsenic compounds in pharmaceutically acceptable form.
  • the arsenic compound in a vial of a kit of the invention may be in the form of a pharmaceutically acceptable solution, e . g . , in combination with sterile saline, dextrose solution, or buffered solution, or other pharmaceutically acceptable sterile fluid.
  • the complex may be lyophilized or desiccated; in this instance, the kit optionally further comprises in a container a pharmaceutically acceptable solution (e.g., saline, dextrose solution, etc.), preferably sterile, to reconstitute the complex to form a solution for injection purposes.
  • kits of the invention further comprises a needle or syringe, preferably packaged in sterile form, for injecting the complex, and/or a packaged alcohol pad. Instructions are optionally included for administration of arsenic compounds by a clinician or by the patient.
  • the magnitude of a therapeutic dose of an arsenic compound in the acute or chronic management of leukemia will vary with the severity of the condition to be treated and the route of administration.
  • the dose, and perhaps dose frequency will also vary according to the age, body weight, condition and response of the individual patient.
  • the daily dose ranges of arsenic trioxide for the conditions described herein are generally from about 0.05 to about 5 mg per kg body weight administered in divided doses administered parenterally or orally or topically.
  • a preferred total daily dose is from about 2.5 to about 40 mg of arsenic trioxide.
  • the arsenic trioxide formulation of the invention is given daily for a maximum of 60 days, or until remission, followed by two to ten additional cycles, each lasting about 25 days in duration.
  • a daily dose of arsenic trioxide greater than or less than 10 mg can be administered.
  • a therapeutic effect can be obtained with a daily dose of arsenic trioxide less than 10 mg.
  • a preferred dosing regimen involves intravenous infusion of about 0.1 to about 5 mg per kg body weight per day for 5 days. This five-day treatment protocol is repeated once per month until the tumor growth tumor is inhibited or when the tumor shows signs of regression.
  • the total daily dose ranges for the conditions described herein are generally from about 0.1 to about 5 mg/kg body weight administered in divided doses administered parenterally or orally or topically.
  • a preferred total daily dose is from about 0.5 to about 4 mg melarsoprol per kg body weight.
  • the effect of the therapy with arsenic trioxide or melarsoprol on development and progression of cancer can be monitored by any methods known in the art, including but not limited to determining: levels of tumor specific antigens and putative biomarkers, e.g., carcinoembryonic antigens (CEA) , alpha-fetoprotein; and changes in morphology and/or size using computed tomographic scan and/or sonogram.
  • CCA carcinoembryonic antigens
  • Desirable blood levels may be maintained by a continuous infusion of an arsenic compound as ascertained by plasma levels. It should be noted that the attending physician would know how to and when to terminate, interrupt or adjust therapy to lower dosage due to toxicity, or bone marrow, liver or kidney dysfunctions. Conversely, the attending physician would also know how to and when to adjust treatment to higher levels if the clinical response is not adequate (precluding toxic side effects) .
  • any suitable route of administration may be employed for providing the patient with an effective dosage of an arsenic compound.
  • oral, transdermal, iontophoretic, parenteral subcutaneous, intramuscular, intrathecal and the like
  • Dosage forms include tablets, troches, cachet, dispersions, suspensions, solutions, capsules, patches, and the like. (See,
  • compositions of the present invention comprise an arsenic compound as the active ingredient, pharmaceutically acceptable salt thereof, and may also contain a pharmaceutically acceptable carrier, and optionally, other therapeutic ingredients, for example all trans retinoic acid.
  • pharmaceutically acceptable salts refers to salts prepared from pharmaceutically acceptable non-toxic acids and bases, including inorganic and organic acids and bases.
  • compositions suitable for oral, mucosal routes, transdermal, iontophoretic, parenteral (including subcutaneous, intramuscular, intrathecal and intravenous) although the most suitable route in any given case will depend on the nature and severity of the condition being treated.
  • a suitable dosage range for use is, e.g.. from about one to about 40 mg arsenic trioxide total daily; about 0.001 to about 10 mg arsenic trioxide per kg body weight total daily, or about 0.1 to about 10 mg melarsoprol per kg body weight total daily.
  • the arsenic carrier could be delivered via charged and uncharged matrices used as drug delivery devices such as cellulose acetate membranes, also through targeted delivery systems such as fusogenic liposomes attached to antibodies or specific antigens.
  • an arsenic compound can be combined as the active ingredient in intimate admixture with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques.
  • the carrier may take a wide variety of forms depending on the form of preparation desired for administration, e.g. , oral or parenteral (including tablets, capsules, powders, intravenous injections or infusions) .
  • any of the usual pharmaceutical media may be employed, e.g. , water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents, and the like; in the case of oral liquid preparations, e.g.
  • compositions for parenteral dosage form such as intravenous injection or infusion
  • similar pharmaceutical media may be employed, e.g. , water, glycols, oils, buffers, sugar, preservatives and the like know to those skilled in the art.
  • parenteral compositions include, but are not limited to Dextrose 5%w/v, normal saline or other solutions.
  • the total dose of the arsenic compound may be administered in a vial of intravenous fluid, e.g. , ranging from about 2 ml to about 2000 ml.
  • the volume of dilution fluid will vary according to the total dose administered.
  • arsenic trioxide supplied as a 10 ml aqueous solution at 1 mg/ml concentration is diluted in 10 to 500 ml of 5% dextrose solution, and used for intravenous infusion over a period of time ranging from about ten minutes to about four hours.
  • An exemplary course of treatment of a patient with leukemia, lymphoma, or solid cancer can involve daily administration by intravenous infusion of arsenic trioxide in an aqueous solution at a daily dose of about 0.01 to 1 mg arsenic trioxide per kg of body weight of the patient. Preferably, about 0.15 mg arsenic trioxide per kg body weight per day is used.
  • the course of treatment may continue until bone marrow remission is observed or when side effects are becoming serious.
  • the course of treatment may be repeated for up to ten times over approximately 10 months with a break of about 3 to 6 weeks in between courses .
  • the post-remission course of treatment involves infusion of arsenic trioxide at a daily dose of about 0.15 mg per kg of body weight of the patient on a daily or weekdays-only basis for a cumulative total of 25 days.
  • arsenic compounds of the invention in treatment of various types of leukemia.
  • the arsenic trioxide formulation of the invention were found to be well- tolerated in humans.
  • three APL patients were given 10 mg of the arsenic trioxide formulation of the invention once daily (flat dose) intravenous dose.
  • the activity of As 2 0 3 and melarsoprol against myeloid leukemia cell lines including the APL cell line NB4- 306 (a retinoic acid-resistant cell line derived from NB4 that no longer expresses the intact PML-RAR ⁇ fusion protein) , HL60, KG-1, and the myelomonocytic cell line U937 was investigated.
  • the inventors also tested these agents using murine embryonic fibroblasts (MEFs) and bone marrow (BM) progenitors in which the PML gene had been inactivated by homologous recombination.
  • MEFs murine embryonic fibroblasts
  • BM bone marrow
  • both As : 0 3 and melarsoprol comparably inhibited growth and induced apoptosis of PML+/+ and PML-/-MEF, and inhibited colony- forming unit erythroid (CFU-E) and CFU granulocyte-monocyte formation in BM cultures of PML+/+ and PML-/- progenitors.
  • CFU-E colony- forming unit erythroid
  • Melarsoprol an organic arsenical synthesized by complexing elarsen oxide with dimercaprol , has primarily been used for the treatment of African trypanosomiasis.
  • the effects of melarsoprol upon induction of apoptosis in cell lines representative of chronic B-cell lymphoproliferative disorders have been investigated, and the results are described below.
  • Melarsoprol (supplied as Arsobal [36 mg/mL] by Rhone Poulenc Rorer, Collegeville, PA) was diluted in propylene glycol at a stock concentration of 10 " " mol/L and stored at room temperature. As 2 0 (Sigma, St. Louis, MO) was dissolved in 1.65 mol/L sodium hydroxide (NaOH) at a stock solution of 10 " - mol/L. Serial dilution (10 "c to 10 " ' mol/L) were made in RPMI 1640 media.
  • An Epstein-Barr virus (EBV)- transformed B-prolymphocytic cell line (JVM-2) , an EBV- transformed B-cell chronic lymphocytic leukemia (B-CLL) cell line (I83CLL) , and one non-EBV-transformed B-CLL cell line (WSU-CLL) were used as targets.
  • EBV Epstein-Barr virus
  • B-CLL B-cell chronic lymphocytic leukemia
  • WSU-CLL non-EBV-transformed B-CLL cell line
  • melarsoprol caused a dose- and time-dependent inhibition of survival and growth in all three cell lines.
  • As ⁇ O at similar concentrations had no effect on either viability or growth.
  • all three cell lines treated with melarsoprol (10 " mol/L) exhibited morphologic characteristics of apoptosis.
  • a prominent concentration-dependent downregulation of bcl-2 RNA after 24 hours of exposure to melarsoprol in WSU-CLL 183CLL, and -TVM-2 cells was observed. Decrease of bcl-2 protein expression was also observed in all three cell lines, whereas As 2 0, had no effect on this parameter.
  • Plasma areas under the concentration x time curves were proportional to the administered dose, ranging from 0.48 ng-hr/ml on Day 1 to 1.48 ng-hr/ml on Day 3. Detectable concentrations of the drug were found in plasma one week after initial dosing. The drug has been relatively well- tolerated. Adverse effects have included transient pain at the injection site and mild nausea. No signs of "reactive encephalopathy" (occasionally observed during treatment of CNS trypanosomiasis) have been observed.
  • APL Acute promyelocytic leukemia
  • RAR ⁇ Retinoic Acid Receptor ⁇
  • transgenic mice were generated by standard techniques in which the expression of the PML-RAR ⁇ or PLZF-RAR ⁇ fusion proteins is placed under the control of a myeloid- promyelocytic specific human Cathepsin-G (hCG) minigene.
  • hCG myeloid- promyelocytic specific human Cathepsin-G
  • hCG-PML/RAR ⁇ and hCG-PLZF-RAR ⁇ transgenic mice develop myeloid leukemia with features of APL similar to those in humans .
  • ATRA prolonged survival, but leukemias relapsed shortly after remission was achieved, and were refractory to further ATRA treatment.
  • the two transgenic mouse models is also used to test the efficacy and dosage of As-0 , and ATRA+As : 0, in combination for treatment of APL patients resistant to ATRA, and in APL associated with the t(ll;17).
  • a regimen of As 2 0- at 6 ⁇ g per day or a combination of As 2 0 : at 6 ⁇ g and ATRA at 1.5 or 6 ⁇ g per gram of body weight per day is administered intraperitoneally . Mice are bled weekly to evaluate the remission of the APL.
  • a 5.0 M HCl solution was prepared by dilution of HCl (49.26 mL, 37% wt/wt, 10/15 M) with H 2 0 (50.74 mL) in a 250 mL Erlenmeyer flask.
  • the HCl solution was later transferred via syringe to a 1000 mL empty evacuated container.
  • the As 2 0 3 solution was back titrated with HCl (0.725 L, 5.0 M) to pH 8.0.
  • Approximately 10 mL of the backtitrated As 2 0, solution was filtered through a Millex-GS 0.22 ⁇ m filter unit and was added to each of approximately 30 sterile evacuated sterile vials.
  • 10 mL of this solution was withdrawn from two of the vials and was added to a 500 mL 5%-dextrose solution which yielded a final pH of 6.5.
  • A-01 Intermediate product solution after initial solubilization in NaOH.
  • A-02 Intermediate product solution prior to HCl titration.
  • A-03 Intermediate product prior to Millex filtration.
  • Arsenic trioxide was evaluated in patients with APL to determine whether this agent induced either cytodifferentiation or apoptosis. Twelve patients who had relapsed from extensive prior therapy were treated with arsenic trioxide at doses ranging from 0.06 to 0.2 mg/kg per day until a bone marrow remission was achieved. Bone marrow mononuclear cells were serially monitored by flow cytometry for immunophenotype, fluorescence in situ hybridization (FISH) , reverse transcription polymerase chain reaction (RT- PCR) assay for PML/RAR- ⁇ expression, and Western blot expression of the apoptosis-associated proteins, caspases 1, 2 and 3.
  • FISH fluorescence in situ hybridization
  • RT- PCR reverse transcription polymerase chain reaction
  • Eligibility requirements included a diagnosis of APL confirmed by cytogenetics or fluorescence in situ hybridization (FISH) analysis for a t(15;17) translocation, or by reverse transcriptase polymerase reaction (RT-PCR) assay for PML/RAR- ⁇ . Patients must have relapsed from standard therapy that had included all-trans retinoic acid plus a combination of cytotoxic drugs. Signed informed consent was required, and the protocol was reviewed and approved by this center's institutional review board
  • Arsenic trioxide treatment Arsenic trioxide was supplied as an aqueous solution in 10 ml vials containing 1 mg/ml of drug. The drug was further diluted in 500 ml of 5%-dextrose solution and infused intravenously over 2 to 4 hours once per day. While the initial cohort of patients received either 10 or 15 g/day as a flat dose, the referral of two children prompted the invention of a weight-based regimen (0.15 mg/kg/day) that was heretofore unknown. The drug was given daily until bone marrow remission was observed. Patients who achieved complete remission were eligible for treatment with additional courses of therapy 3 to 6 weeks after the preceding course.
  • Subsequent courses were generally given at a dose of 0.15 mg/kg/day for a cumulative total of 25 days, administered either daily or on a weekdays-only schedule, for a maximum total of 6 courses over approximately 10 months.
  • Patients with coagulopathy were transfused with platelets and fresh-frozen plasma to maintain the platelet count and fibrinogen at target levels > 50,000 cells/cu mm and > 100 mg/dL, respectively.
  • Blood counts, coagulation studies, serum chemistry profiles, urinalyses, and electrocardiograms were serially obtained.
  • a bone marrow aspiration and/or biopsy was performed at baseline and periodically thereafter until remission was documented. Conventional response criteria were observed, which included recovery of bone marrow to ⁇ . 5% blasts, peripheral blood leukocytes > 3,000 cells/cu mm, and platelets > 100,000 cells/cu mm.
  • Fluorescence in situ hybridization Selected specimens that had undergone immunofluorescence staining for CD33 and CDllb were sorted for cells that coexpressed both antigens using a FACStar Plus cell-sorter (Becton-Dickinson) . Separated cells were incubated in culture media at 37° C for one hour, treated with hypotonic solution 0.075M KC1 for 5 minutes, fixed in 3 : 1 methanol: acetic acid fixative, and air- dried. Interphase FISH was performed using a specific
  • PML/RAR- ⁇ translocation dual-color probe (Vysis; Downer's Grove, IL) . Briefly, DNA from interphase cells was denatured by immersing slides in a solution of 50% formamide/2xSSC at 73° C for 5 minutes; the slides were then dehydrated in alcohol and air dried. A mixture of probe in hybridization mixture was applied, covered with a cover slip, and sealed with rubber cement. Hybridization was carried out at 37° C in a moist chamber for approximately 12 to 16 hours. Following hybridization, unbound probe was removed by washing the slides at 45° C in 50% formamide/2xSSC solution three times for 10 minutes each, followed by a wash in 2xSSC/0.1 NP-40 solution at 45° C for 5 minutes. Slides were then air dried and counter-stained with 4 ' , 6-diamidino-2-phenylindole and covered with a glass coverslip. Analysis of interphase cells for fluorescent signals was performed with a
  • Photometries Sensys camera fitted to a Zeiss axioscope A minimum of 300 cells was studied for each sample.
  • RT-PCR analysis for PML/RAR- ⁇ was performed using methods previously described (Miller et al., 1992, Proc. Natl. Acad. Sci. 89:2694-8; Miller et al., 1993, Blood, 82:1689-94) .
  • RESULTS Patients Twelve patients with relapsed or refractory APL were treated. All patients had received extensive prior therapy with retinoids and cytotoxic drugs (Table 3). Two patients had relapsed from allogeneic bone marrow transplantation, one of whom had also failed donor T-cell reinfusion. One patient was being maintained on hemodialysis for chronic renal failure.
  • Clinical Efficacy Eleven of the 12 patients achieved a complete remission after arsenic trioxide treatment.
  • the patient who entered on hemodialysis sustained an intracranial hemorrhage on day 1 and died on day 5.
  • the median duration of therapy in responding patients was 33 days (range, 12 to 39 days), the median daily dose was 0.16 mg/kg (range, 0.06 to 0.2 mg/kg), and the median cumulative dose during induction was 360 mg (range, 160 to 515 mg) (Table 3) .
  • Complete remission by all criteria was attained at a median time of 47 days (range, 24 to 83 days) after initiation of therapy.
  • All 11 patients in complete remission completed at least 1 post-remission treatment course with arsenic trioxide.
  • the median duration of remission is 5+ months (range, 1 to 9+ months) .
  • 3 of the 11 patients relapsed during the second treatment course; none of these patients had converted their RT-PCR assays, and each appeared to have rapidly acquired drug resistance. Two of these individuals have since expired from progressive leukemia.
  • Adverse Events The clinical condition of patients in this study was highly variable, which reflected their extensive prior therapy. The protocol did not require hospitalization; three patients completed induction therapy entirely as outpatients, and one other individual was hospitalized solely for placement of a venous catheter. However, 8 patients were hospitalized for complications of leukemia, 5 of whom required transfer to an intensive care unit, endotracheal intubation, and assisted ventilation for complications that included pulmonary hemorrhage, renal failure, sepsis, graft vs. host disease, non-specific pulmonary infiltrates, or hypotension. One patient required insertion of a permanent pacemaker after second-degree heart block developed in the setting of severe metabolic acidosis, hyperkalemia, hypotension, and renal insufficiency.
  • the median total peripheral blood leukocyte count at entry was 4,700 cells/cu mm (range, 500 to 144,000 cells/cu mm).
  • Six patients developed leukocytosis (i.e., > 20,000 cells/cu mm) that ranged from 20,800 to 144,200 cells/cu mm. No additional therapy was administered to these patients, and the leukocytosis resolved in all cases without further intervention.
  • APL is characterized by cells that express CD33, an antigen typically associated with primitive myeloid cells.
  • Arsenic trioxide therapy induced a progressive decrease in the proportion of cells that solely expressed CD33, along with an increase in the proportion of cells that expressed CDllb, an antigen associated with mature myeloid elements. While these changes would be anticipated from any agent that induced remission in APL, arsenic trioxide also induced expression of cells that simultaneously expressed both antigens. In most cases, these dual- expressing cells dominated the myeloid cell population, and they persisted for extended periods after complete remission was achieved by clinical criteria.
  • Fluorescence in situ hybridization analysis Bone marrow mononuclear cells taken from a patient both early and later in complete remission were sorted by flow cytometry for coexpression of CD33 and CDllb. Using fluorescence in situ hybridization (FISH) analysis, three hundred cells were examined early in remission. Similar to control APL cells, the majority of these cells yielded a hybrid signal, indicating a translocation between PML and RAR- ⁇ genes and their origin from the neoplastic clone. However, when cells from the same patient were again sorted using these same parameters later in remission, only the normal pattern of fluorescence signals was detected, indicating their derivation from normal hematopoietic progenitors.
  • FISH fluorescence in situ hybridization
  • patients admitted to the trial had sustained multiple relapses and were resistant to conventional chemotherapy, retinoids, or bone marrow transplantation.
  • patients in this study suffered from numerous leukemia-related complications, including respiratory failure, disseminated Varicella zoster infection, cavitary aspergillosis, chronic renal failure, and graft-vs. -host disease.
  • 5 of the 12 patients required admission to an intensive care unit for assisted ventilation and supportive care, but these complications were not directly related to arsenic trioxide therapy.
  • All-trans retinoic acid induces "terminal" differentiation of APL cells, but the cytodifferentiating effects of arsenic trioxide appear to be incomplete.
  • Arsenic induces a population of cells that simultaneously express surface antigens characteristic of both mature and immature cells (i.e. CDllb and CD33, respectively). Early during induction, these cells retain the t(15;17) translocation that characterizes APL. Unexpectedly, these cells persisted in the bone marrow despite the achievement of a clinically complete remission; however, later in remission, the coexpressing cells - while still readily detectable - were no longer positive by in situ hybridization.
  • leukemic cells during therapy are also far less distinctive than that observed during therapy with all-trans retinoic acid.
  • leukemic cells from many patients ⁇ isplayed few morphologic changes for 10 or more days, after which the proportion of leukemic cells progressively decreased.
  • arsenic trioxide appeared to induce apoptosis, coincident with increased expression and conversion of cysteine proteases (termed caspases) from inactive precursors to activated enzymes.
  • the caspase pathway has only recently been characterized as an important pathway of programmed cell death. Initially recognized due to homology between the C.
  • caspase activation is inducible with a number of cytotoxic agents, including all-trans retinoic acid. Since these enzymes induce widespread proteolysis, it is conceivable that PML/RAR- ⁇ is a caspase substrate.
  • a final similarity shared by arsenic trioxide and all-trans retinoic acid is the rapid development of clinical resistance in some individuals.
  • arsenic trioxide induces complete remission in patients with APL who have relapsed from extensive prior therapy.
  • This drug causes partial but incomplete cytodifferentiation of leukemic cells, followed by caspase activation and induction of apoptosis.
  • Table 3 Clinical characteristics and induction therapy results of patients with acute
  • the inventors Based upon the initial discovery of the antitumor effects of arsenic trioxide in vitro against B-cell lymphocyte lines, the inventors treated one patient with intermediate-grade large cell lymphoma who had relapsed from multiple forms of conventional therapy, including autologous bone marrow transplantation. Despite rapid progression of his disease prior to starting the arsenic trioxide therapy, treatment with arsenic trioxide effected a major (>50%) shrinkage in the size of his cancerous lymph nodes and spleen, which was also associated with a major improvement of his quality of life.
  • PK pharmacokinetics
  • FISH fluorescence in situ hybridization

Abstract

The invention relates to the use of arsenic compounds to treat a variety of leukemia, lymphoma and solid tumors. Further, the arsenic compounds may be used in combination with other therapeutic agents, such as a retinoid. The invention also provides a process for producing arsenic trioxide formulations.

Description

PROCESS FOR PRODUCING
ARSENIC TRIOXIDE FORMULATIONS AND
METHODS FOR TREATING CANCER
USING ARSENIC TRIOXIDE OR MELARSOPROL
1. FIELD OF INVENTION
The present invention relates to methods and compositions for the treatment of leukemia, lymphoma, and certain other cancers.
More specifically, the present invention relates to the novel uses of arsenic trioxide and an organic arsenic compound for treating acute leukemia and chronic leukemia.
2. BACKGROUND OF THE INVENTION
2.1. CANCER
Cancer is characterized primarily by an increase in the number of abnormal cells derived from a given normal tissue, invasion of adjacent tissues by these abnormal cells, and lymphatic or blood-borne spread of malignant cells to regional lymph nodes and to distant sites (metastasis) . Clinical data and molecular biologic studies indicate that cancer is a multistep process that begins with minor preneoplastic changes, which may under certain conditions progress to neoplasia.
Pre-malignant abnormal cell growth as exemplified by hyperplasia, metaplasia, and dysplasia (for review of such abnormal growth conditions, see Robbins and Angell, 1976, Basic Pathology, 2d Ed., W.B. Saunders Co., Philadelphia, pp. 68-79) precedes the formation of a neoplastic lesion. A neoplastic lesion may evolve clonally to grow into a solid tumor, and develop an increasing capacity for invasion, growth, metastasis, and heterogeneity, especially under conditions in which the neoplastic cells escape the host's immune surveillance (Roitt, I., Brostoff, J and Kale, D. , 1993, Immunology, 3rd ed. , Mosby, St. Louis, pps. 17.1- 17.12) . Leukemia refers to malignant neoplasms of the blood-forming tissues. Transformation to malignancy typically occurs in a single cell through two or more steps with subsequent proliferation and clonal expansion. In some leukemias, specific chromosomal translocations have been identified with consistent leukemic cell morphology and special clinical features (e.g., translocations of 9 and 22 in chronic myelocytic leukemia, and of 15 and 17 in acute promyelocytic leukemia) . Acute leukemias are predominantly undifferentiated cell populations and chronic leukemias more mature cell forms.
Acute leukemias are divided into lymphoblastic (ALL) and non-lymphoblastic (ANLL) types. They may be further subdivided by their morphologic and cytochemical appearance according to the French-American-British (FAB) classification or according to their type and degree of differentiation. The use of specific B- and T-cell and myeloid-antigen monoclonal antibodies are most helpful for classification. ALL is predominantly a childhood disease which is established by laboratory findings and bone marrow examination. ANLL, also known as acute myeloblastic leukemia (AML) , occurs at all ages and is the more common acute leukemia among adults; it is the form usually associated with irradiation as a causative agent. Chronic leukemias are described as being lymphocytic (CLL) or myelocytic (CML) . CLL is characterized by the appearance of mature lymphocytes in blood, bone marrow, and lymphoid organs. The hallmark of CLL is sustained, absolute lymphocytosis (> 5,000/μL) and an increase of lymphocytes in the bone marrow. Most CLL patients also have clonal expansion of lymphocytes with B- cell characteristics. CLL is a disease of older persons. In CML, the characteristic feature is the predominance of granulocytic cells of all stages of differentiation in blood, bone marrow, liver, spleen, and other organs. In the symptomatic patient at diagnosis the total BC count is usually about 200,000/μL, but may reach 1, 000, 000/μL. CML is relatively easy to diagnose because of the presence of the Philadelphia chromosome.
The very nature of hematopoietic cancer necessitates using systemic chemotherapy as the primary treatment modality. Drugs selected according to sensitivities of specific leukemias are usually given in combination. Radiation therapy may be used as an adjunct to treat local accumulations of leukemic cells. Surgery is rarely indicated as a primary treatment modality, but may be used in managing some complications. Bone marrow transplantation from an HLA-matched sibling is sometimes indicated.
2.2. ARSENIC AND ITS MEDICAL USES Arsenic has been considered to be both a poison and a drug for a long time in both Western and Chinese medical practices. In the latter part of the nineteenth century, arsenic was used frequently in attempts to treat diseases of the blood in the West. In 1878, it was reported that treatment of a leukemic patient with Fowler's solution (a solution containing potassium arsenite, valence +5) reduced markedly the count of white blood cells (Cutler and Bradford, Am. J. Med. Sci.. January 1878, 81-84). Further interests in the use of Fowler's solution as a palliative agent to treat chronic myelogenous leukemia (CML) was described by Forkner and Scott in 1931 (J. Am. Med. Assoc. , 1931, iii, 97) , and later confirmed by Stephens and Lawrence in 1936 (Ann. Intern. Med. 9, 1488-1502). However, while the active chemical ingredient (s) of Fowler's solution was not determined, its toxicity was well recognized. Fowler's solution was administered strictly as an oral composition, and was given to leukemic patients as a solution until the level of white blood cells was depressed to an acceptable level or until toxicities (such as skin keratoses and hyperpigmentation) developed, while the patients enjoyed varying periods of remission. In the 1960 's, Fowler's solution was still used occasionally in attempts to treat CML, however, most patients with CML were treated with other chemotherapeutic agents, such as busulfan, and/or radiation therapy (Monfardini et al . , Cancer. 1973, 31:492-501).
Paradoxically, one of the long recognized effects of exposure to arsenic, whether the source is environmental or medicinal, is skin cancer (Hutchinson, 1888, Trans. Path. Soc. Lond.. 39:352; Neubauer, 1947, Br. J. Cancer. 1:192). There were even epidemiological data to suggest that the use of Fowler's solution over long periods could lead to an increased incidence of cancer at internal sites (Cuzick et al . , Br. J. Cancer. 1982, 45:904-911; Kaspar et al . , J. Am.
Med. Assoc.. 1984, 252:3407-3408). The carcinogenicity of arsenic has since been demonstrated by the fact that it can induce chromosomal aberration, gene amplification, sister chromatid exchanges and cellular transformation (See e.g., Lee et al . , 1988, Science, 241:79-81; and Germolec et al . ,
Toxicol. Applied Pharmacol.. 1996, 141:308-318). Because of the known carcinogenic effect of arsenic, its only therapeutic use in human in Western medicine today is in the treatment of tropical diseases, such as African trypanosomiasis, (the organic arsenical, melarsoprol; See
Goodman & Gil an's The Pharmacological Basis of Therapeutics, 9th edition, chapter 66, 1659-1662, 1997).
In traditional Chinese medicine, arsenous acid or arsenic trioxide paste has been used to treat tooth marrow diseases, psoriasis, syphilis and rheumatosis (Chen et al . ,
1995, in Manual of Clinical Drugs, Shanghai, China, Shanghai Institute of Science and Technology, p.830). In 1970's, arsenic trioxide had been applied experimentally to treat acute promyelocytic leukemia (APL) in China (commented by Mervis, 1996, Science. 273:578). The clinical efficacy of arsenic trioxide has recently been re-investigated in 14 of 15 patients with refractory APL, where the use of an intravenous dose at 10 mg/day for 4-9 weeks was reported to result in complete morphologic remission without associated bone marrow suppression (Shen et al . , 1997, Blood, 89:3354-
3360) . It was also shown that arsenic trioxide induced apoptosis (programmed cell death) iri vitro in NB4 cells, an APL cell line, and that apoptosis was apparently associated with down-regulation of the oncogene bcl-2 , and intracellular redistribution of the chimeric PML/RARα protein that are unique to APL cells (Chen et al . , 1996, Blood, 88:1052-1061; Andre et al . , 1996, Exp. Cell Res. 229:253-260). It has been reported that the biological activity of arsenic is due to the ability of arsenic to direct the nucleoplasmic fraction of PML to nuclear bodies for degradation (Zhu et al., 1997, Proc. Natl. Acad. Sci., 94:3978-3983). Although arsenic is well known to be both a poison and a carcinogenic agent, there have been many reports concerning the use of arsenic in medical treatment. Further, from the above discussion, it should be clear that there are many different types of leukemias, each of which requires a unique treatment protocol that is modified according to the presence of factors predicting for a risk of treatment failure. Thus, the development of a broad spectrum anti- leukemia agent that can be used alone or in combination with other existing drugs is extremely desirable.
3. SUMMARY OF THE INVENTION
Despite the conflicting reports in the art concerning benefits and risks of the administration of arsenic to patients, applicants have discovered that arsenic rioxide and the organic arsenical, melarsoprol, have broad applicability in the treatment of various types of leukemias, lymphomas, and solid tumors.
The invention described herein encompasses a method of treating leukemia, lymphoma or solid tumors comprising the administration of a therapeutically effective and non-lethal amount of arsenic trioxide or melarsoprol to a human in need of such therapy. The invention, as mentioned above also encompasses the use of combination therapy to treat leukemia, especially leukemias which are refractory to other forms of treatment. The invention also encompasses a method for the manufacture of pharmaceutical compositions comprising arsenic trioxide.
In accordance with the present invention, arsenic trioxide or melarsoprol compounds can be used alone or in combination with other known therapeutic agents (including chemotherapeutics, radioprotectants and radiotherapeutics) or techniques to either improve the quality of life of the patient, or to treat leukemia, lymphoma or solid tumor. The arsenic compounds can be used before, during or after the administration of one or more known chemotherapeutic agents, including antitumor agents. In addition, the arsenic compounds can be used before, during or after radiation treatment. The pharmaceutical compositions of the invention are sterile solutions suitable for intravenous injection or infusion. In another embodiment the invention encompasses a composition suitable for oral delivery; comprising arsenic trioxide or melarsoprol and a pharmaceutically acceptable excipient or carrier. In another embodiment, the invention also includes compositions suitable for topical or transdermal delivery, including but not limited to iontophoretic methods. Specific therapeutic regimens, pharmaceutical compositions, and kits are also provided by the invention.
Particular compositions of the invention and their uses are described in the sections and subsections which follow.
4. DETAILED DESCRIPTION OF THE INVENTION
Methods and compositions for the treatment of leukemia, lymphoma or solid tumors are described herein. This invention provides a method of treating acute or chronic leukemia, lymphoma, or solid tumors in a human which comprises administering to a human in need of such therapy a therapeutically effective and non-lethal amount of one or more arsenic compounds, such as arsenic trioxide or melarsoprol. The invention also includes a method of treating leukemia in a human who has become refractory to other forms of treatment which comprises administering to a human arsenic trioxide or melarsoprol in combination with another chemotherapeutic agent, e.g., all-trans retinoic acid (ATRA) .
The invention also relates to a method for the manufacture of pharmaceutical compositions comprising arsenic trioxide. It is preferred that pharmaceutical compositions of the present invention exhibit reduced toxicity, improved efficacy, improved stability during storage and use, and that the composition has a physiologically acceptable pH.
4.1. THE ARSENIC COMPOUNDS
As used herein, "arsenic compound" refers to a pharmaceutically acceptable form of arsenic trioxide (As03) or melarsoprol. Melarsoprol is an organic arsenic compound which can be synthesized by complexing melarsen oxide with dimercaprol or commercially purchased (Arsobal® by Rhone Poulenc Rorer, Collegeville, PA) . Since the non- pharmaceutically formulated raw materials of the invention are well known, they can be prepared from well-known chemical techniques in the art. (See for example , Kirk-Othmer,
Encyclopedia of Chemical Technology 4th ed. volume 3 pps. 633-655 John Wiley & Sons) . As used herein the terms "a therapeutic agent",
"therapeutic regimen", "radioprotectant" , "chemotherapeutic" mean conventional drugs and drug therapies, including vaccines, for treating cancer, viral infections, and other malignancies, which are known to those skilled in the art. "Radiotherapeutic" agents are well known in the art.
In accordance with the present invention, arsenic trioxide or melarsoprol compounds can be used alone or in combination with other known therapeutic agents (including chemotherapeutics, radioprotectants and radiotherapeutics) or techniques to either improve the quality of life of the patient, or to treat leukemia, lymphoma or solid tumor. For example, the arsenic compounds can be used before, during or after the administration of one or more known antitumor agents including but not limited to mustard compounds, nitrogen mustard, chlorambucil, melphalan, cyclophosphamide, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil, floxuridine, methotrexate, vincristine, vinblastine, taxol, etoposide, temiposide, dactinomycin, daunorubicin, doxorubicin, bleomycin, mitomycin, cisplatin, carboplatin, estramustine phosphate, hydroxyurea, BCNU, procarbazine, VM- 26, interferons, and all-trans retinoic acid (ATRA) , or other retinoids (See, for example , the Physician Desk References 1997) . In addition, the arsenic compounds can be used before, during or after radiation treatment.
In a specific embodiment, the arsenic compound of the invention and ATRA can be administered as a mixture. In preferred aspects, the lymphoma, leukemia or solid tumor in the human treated by the combination is refractory to general methods of treatment, or is a relapsed case of leukemia.
Any suitable mode of administration may be used in accordance with the present invention including but not limited to parenteral administration such as intravenous, subcutaneous, intramuscular and intrathecal administration; oral, and intranasal administration, and inhalation. The mode of administration will vary according to the type of cancer, and the condition of the human.
The pharmaceutical compositions to be used may be in the form of sterile aqueous or organic solutions, colloidal suspensions, caplets, tablets and cachets.
4.2. METHODS OF TREATMENT
The term "a method for treating leukemia" as used herein means that the disease and the symptoms associated with the disease are alleviated, reduced, cured, or placed in a state of remission. For example, the methods of treatment of the invention can lower the white blood cell count, or reduce lymphocytosis in a human under treatment. The term "a method for treating lymphoma" as used herein means that the disease and the symptoms associated with the disease are alleviated, reduced, cured, or placed in a state of remission. The term "a method for treating solid tumor" as used herein means that the disease and the symptoms associated with the solid tumor are alleviated, reduced, cured, or placed in a state of remission. In addition, the term "a method for treating leukemic infiltration" means that the infiltration of leukemic cells out of circulation and into other organs and systems and the symptoms associated with such infiltration are alleviated, reduced, cured, or placed in a state of remission.
The term "refractory" when used herein means that the leukemia is generally resistant to treatment or cure.
As used herein, "preneoplastic" cell refers to a cell which is in transition from a normal to a neoplastic form; or cells that fail to differentiate normally; and morphological evidence, increasingly supported by molecular biologic studies, indicates that preneoplasia progresses through multiple steps.
In one embodiment, the invention provides a method for treatment of leukemia in a human comprising the administration of a therapeutically effective and non-lethal amount of arsenic trioxide or melarsoprol to the human. The invention also provides a weight-based dosing regimen, not heretofore disclosed, that maximizes the safety in humans of these otherwise highly toxic compounds.
Arsenic trioxide (As:0. ) inhibits growth and induce apoptosis in NB4 acute promyelocytic leukemic cells. Acute promyelocytic leukemia (APL) is associated with the t(15;17) translocation, which generates a PML/RAR fusion protein between PML, a growth suppressor localized on nuclear matrix- associated bodies, and RARα, a nuclear receptor for retinoic acid (RA) . PML/RARα was proposed to block myeloid differentiation through inhibition of nuclear receptor response, as does a dominant negative RARα mutant. In addition, in APL cells, PML/RARα displaces PML and other nuclear body (NB) antigens onto nuclear microspeckles, likely resulting in the loss of PML and/or NB functions. It has been suggested that high concentrations of arsenic trioxide promote apoptosis, whereas low concentrations induce partial differentiation in NB4 cells as well as cells derived from APL patients. It was postulated that As203 works through its ability to specifically cause PML-RARα in APL cells to be relocalized to nuclear bodies for degradation (Zhu et al. , 1997, Proc. Natl. Acad. Sci . USA. 94:3978-3983). However, these findings tend to limit the use of arsenic trioxide to a subset of leukemias. See Konig et al. , 1997, Blood, 90:562- 570. Unexpectedly, the inventors have discovered that both As20; and melarsoprol are able to inhibit cell growth, and induce apoptosis in various myeloid leukemia cell lines in a PML and PML-RARα independent manner. Thus, the inventors have discovered that, contrary to the earlier findings, arsenic trioxide and melarsoprol are both effective against a broad range of leukemias regardless of the underlying molecular mechanism that causes the neoplasia. Working examples of the effect of arsenic compounds on a number of leukemic cell lines are provided in Sections 5.1 and 5.2.
Accordingly, the arsenic compounds of the present invention can be used against a variety of leukemias, including but not limited to:
Acute lymphoblastic leukemia (ALL) Acute lymphoblastic B-cell leukemia
Acute lymphoblastic T-cell leukemia
Acute yeloblastic leukemia (AML)
Acute promyelocytic leukemia (APL)
Acute monoblastic leukemia Acute erythroleukemic leukemia
Acute megakaryoblastic leukemia
Acute myelomonocytic leukemia
Acute undifferentiated leukemia
Chronic myelocytic leukemia (CML) Chronic lymphocytic leukemia (CLL)
The skilled artisan will recognize that other leukemias may be treated in accordance with the present invention. In another embodiment, the invention provides a method for treatment of lymphoma in a human comprising the administration of a therapeutically effective and non-lethal amount of arsenic trioxide or melarsoprol to the human. Lymphoma that can be treated by the methods of the invention include but are not limited to high grade lymphoma, intermediate grade lymphoma, low grade lymphoma, and the various subclassifications.
In yet another embodiment, the invention provides a method for treatment of solid tumors, including metastasises, in humans comprising the administration of a therapeutically effective and non-lethal amount of arsenic trioxide or melarsoprol to the human. Solid tumors that can be treated by the methods of the invention include but are not limited to: cancer of the digestive tract, oesophagus, liver, stomach, and colon; skin; brain; bone; breast; lung; and soft tissues, including but not limited to various sarcomas, and preferably prostate cancer.
In various embodiments, the leukemic or tumor cells are infiltrating other organs and systems in a human, for example, the central nervous system. The methods of the invention are also applicable to reduce the number of preneoplastic cells in a human in which there is an abnormal increase in the number of preneoplastic cells. In a specific embodiment, the invention provides a method of treatment of acute promyelolytic leukemia (APL) in a human comprising the administration of a therapeutically effective and non-lethal amount of melarsoprol to the human. The inventors discovered, as described in Section 5.2, that concentrations of melarsoprol that are cytotoxic in vitro can readily be achieved in vivo .
In one specific embodiment, the invention provides a method of treatment of chronic myelogenous leukemia (CML) in a human comprising the administration of a therapeutically effective and non-lethal amount of arsenic trioxide to the human. The inventors discovered, as described in Section 5.3, that arsenic trioxide can also induce apoptosis in a CML cell line. The therapeutic benefits of the pharmaceutical compositions of the invention comprising arsenic trioxide is far superior to that of potassium arsenite, commonly formulated as Fowler's solution.
In yet another specific embodiment, the invention provides a method of treatment of acute promyelocytic leukemia (APL) in a human, in which the APL is associated with a translocation of the RARα locus on chromosome 17 to chromosome 11, comprising the administration of a therapeutically effective amount of arsenic trioxide or melarsoprol to the human. In the majority of APL cases, RARα on chromosome 17 translocates and fuses with the PML gene located on chromosome 15, i.e., t(15;l7). In a few cases RARα translocates to chromosome 11 where it fuses to the PLZF gene. Patients harboring the t(l5;17) are uniquely sensitive to treatment with all-trans retinoic acid (ATRA) , yielding complete remission rates of 75% to 95%. APL associated with the t(ll;17) (PLZF-RARα) shows a distinctly worse prognosis with poor response to chemotherapy and little or no response to treatment with ATRA, thus defining a new APL syndrome. The present invention provides that arsenic trioxide or melarsoprol can be used to treat such cases of APL. Transgenic animal models of APL associated with t(15;17) and t(ll;17) for testing the therapeutic benefits and dosages of arsenic compounds of the invention are described in Section 5.4 hereinbelow.
Humans having leukemia are sometimes refractory to conventional methods of treatment by reason of having undergone anti-leukemic therapy (e . g . , chemotherapy). Thus, the invention provides a method of treatment of leukemia in a human who is not responding to conventional therapy comprising the administration of a therapeutically effective and non-lethal amount of a combination of arsenic compound and another chemotherapeutic agent, such as but not limited to, all-trans retinoic acid (ATRA) or other retinoids, to the human. The arsenic compound can either be arsenic trioxide or melarsoprol or a pharmaceutically acceptable salt thereof. The invention also encompasses the treatment of retinoid- resistant patients with an arsenic compound. In specific embodiments, the arsenic compound of the invention and the chemotherapeutic agent can be administered either as a mixture or sequentially. When administered sequentially, the arsenic compound may be administered before or after the chemotherapeutic agent, so long as the first administered agent is still providing antileukemic activity in the human when the second agent is administered. Any of the modes of administration described herein may be used to deliver the combination. In preferred aspects, the leukemia in the human treated by the combination is refractory to general methods of treatment, or is a relapsed case of leukemia.
4.3. PROCESS FOR THE MANUFACTURE OF STERILE ARSENIC TRIOXIDE SOLUTION
The arsenic compounds of the invention may be formulated into sterile pharmaceutical preparations for administration to humans for treatment of leukemias, lymphomas and solid tumors. Compositions comprising a compound of the invention formulated in a compatible pharmaceutical carrier may be prepared, packaged, labelled for treatment of and used for the treatment of the indicated leukemia, lymphoma, or solid tumor.
In one aspect, the invention provides a method for the manufacture of a pharmaceutical composition comprising a therapeutic effective and non-lethal amount of arsenic trioxide (As:03) . Arsenic trioxide (raw material) is a solid inorganic compound that is commercially available in a very pure form. However, it is difficult to dissolve As:0, in aqueous solution. Further, the inventors are unaware of any published teachings on how to formulate As:0, as a pharmaceutical composition suitable for injection directly into a human. Arsenic is present in solution in the +5 valence state (pentavalent) or the +3 valence state (trivalent) . For example, potassium arsenite (KAsO, ; which is present in Fowler's solution) and salts of arsenious acid contain pentavalent arsenic. It is known that one form of arsenic is more toxic than the other. (Goodman & Gilman's The Pharmacological Basis of Therapeutics, 9th edition, chapter 66, 1660, 1997) . A fresh solution of arsenic trioxide containing arsenic in the trivalent state will be gradually oxidized to pentavalent state if exposed to air for a prolonged period, and as a result of the accumulation of pentavalent arsenic, the relative toxicity of a solution of As:03 will change over time. (Id . ) Furthermore, it is observed that the total amount of arsenic in solution decreases over time. This loss of material is caused by the progressive conversion of arsenic in the solution into arsine (AsH;. ) which is a gaseous compound at room temperature. This is particularly problematic in pharmaceutical applications if the concentration of an active ingredient in the injected material cannot be controlled. It is also undesirable to allow arsine to escape from the solution into the atmosphere because arsine is also toxic.
The inventors have experimented and successfully developed a method for formulating arsenic trioxide which overcomes the above-described problems of solubility and stability. The method comprises solubilizing solid high purity As:0? in an aqueous solution at high pH, such as pH greater than 12. For example, a 5 M solution of sodium hydroxide may be used. To aid solubilization and obtain a clear and homogenous solution, mechanical stirring and/or gentle heating may be applied. A solution of As20. can also be obtained by dissolving the solid compound overnight. Typically, a solution of 1 M As:0. is obtained by this method. However, this solution is too basic to be useful as a pharmaceutical composition.
To adjust the pH of the As-,0, solution, the solution is first diluted in water, for example, to a concentration of about 1 mg/mL, pH 12. The As:0. solution is then back- titrated with acid, such as, hydrochloric acid (1 M to 5 M HC1) , with constant stirring until the pH is about 8.0 to 8.5. Highly concentrated HCl is not suitab.le as it causes precipitation to occur in the As0;. solution. The partially neutralized As-0? solution may then be sterilized for example, by filtration (e.g., through a 0.22 μm filter), and stored in sterile vials.
To make a pharmaceutical composition that can be directly injected into a subject, the composition must be sterile, standard techniques known to the skilled artisan for sterilization can be used. See, e.g., Remington's
Pharmaceutical Science, the pH of the partially neutralized As:0. solution may be further adjusted to near physiological pH by dilution (10-100 fold) with a pharmaceutical carrier, such as a 5% dextrose solution. For example, lOmL of a partially neutralized As:0 solution can be added to 500 mL of a 5% dextrose solution to yield a final pH of about 6.5 to 7.5. The method of the invention reduces the oxidation of arsenic in solution. Pharmaceutical compositions containing arsenic trioxide manufactured by the method of the invention show improved stability and long shelf life.
4.4. PHARMACEUTICAL COMPOSITION AND MODES OF ADMINISTRATION
According to the invention, the arsenic compounds and their physiologically acceptable solvates may be formulated for oral or parenteral administration.
For oral administration, the pharmaceutical preparation may be in liquid form, for example, solutions, syrups or suspensions, or may be presented as a drug product for reconstitution with water or other suitable vehicle before use. Such liquid preparations may be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e.g., sorbitol syrup, cellulose derivatives or hydrogenated edible fats) ; emulsifying agents (e .g . , lecithin or acacia); non-aqueous vehicles (e . g. , almond oil, oily esters, or fractionated vegetable oils) ; and preservatives (e . g . , methyl or propyl-p-hydroxybenzoates or sorbic acid) . The pharmaceutical compositions may take the form of, for example, tablets or capsules prepared by conventional means with pharmaceutically acceptable excipients such as binding agents (e . g . , pregelatinized maize starch, polyvinyl pyrrolidone or hydroxypropyl methylcellulose) ; fillers (e.g., lactose, microcrystalline cellulose or calcium hydrogen phosphate); lubricants (e.g., magnesium stearate, talc or silica); disintegrants (e.g., potato starch or sodium starch glycolate) ; or wetting agents (e.g., sodium lauryl sulphate). The tablets may be coated by methods well-known in the art.
For administration by inhalation, the compounds for use according to the present invention are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of, e .g. , gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
The compounds may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampules or in multi- dose containers, with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use. The invention also provides kits for carrying out the therapeutic regimens of the invention. Such kits comprise in one or more containers therapeutically effective amounts of the arsenic compounds in pharmaceutically acceptable form. The arsenic compound in a vial of a kit of the invention may be in the form of a pharmaceutically acceptable solution, e . g . , in combination with sterile saline, dextrose solution, or buffered solution, or other pharmaceutically acceptable sterile fluid. Alternatively, the complex may be lyophilized or desiccated; in this instance, the kit optionally further comprises in a container a pharmaceutically acceptable solution (e.g., saline, dextrose solution, etc.), preferably sterile, to reconstitute the complex to form a solution for injection purposes.
In another embodiment, a kit of the invention further comprises a needle or syringe, preferably packaged in sterile form, for injecting the complex, and/or a packaged alcohol pad. Instructions are optionally included for administration of arsenic compounds by a clinician or by the patient.
The magnitude of a therapeutic dose of an arsenic compound in the acute or chronic management of leukemia will vary with the severity of the condition to be treated and the route of administration. The dose, and perhaps dose frequency, will also vary according to the age, body weight, condition and response of the individual patient. In general, the daily dose ranges of arsenic trioxide for the conditions described herein are generally from about 0.05 to about 5 mg per kg body weight administered in divided doses administered parenterally or orally or topically. A preferred total daily dose is from about 2.5 to about 40 mg of arsenic trioxide. Preferably the arsenic trioxide formulation of the invention is given daily for a maximum of 60 days, or until remission, followed by two to ten additional cycles, each lasting about 25 days in duration. For example, depending on the body weight of a patient with acute promyelocytic leukemia, a daily dose of arsenic trioxide greater than or less than 10 mg can be administered. Alternatively, following the weight-based dosing regimen, a therapeutic effect can be obtained with a daily dose of arsenic trioxide less than 10 mg. For treatment of solid tumor, a preferred dosing regimen involves intravenous infusion of about 0.1 to about 5 mg per kg body weight per day for 5 days. This five-day treatment protocol is repeated once per month until the tumor growth tumor is inhibited or when the tumor shows signs of regression.
As for melarsoprol, the total daily dose ranges for the conditions described herein are generally from about 0.1 to about 5 mg/kg body weight administered in divided doses administered parenterally or orally or topically. A preferred total daily dose is from about 0.5 to about 4 mg melarsoprol per kg body weight.
The effect of the therapy with arsenic trioxide or melarsoprol on development and progression of cancer can be monitored by any methods known in the art, including but not limited to determining: levels of tumor specific antigens and putative biomarkers, e.g., carcinoembryonic antigens (CEA) , alpha-fetoprotein; and changes in morphology and/or size using computed tomographic scan and/or sonogram.
Desirable blood levels may be maintained by a continuous infusion of an arsenic compound as ascertained by plasma levels. It should be noted that the attending physician would know how to and when to terminate, interrupt or adjust therapy to lower dosage due to toxicity, or bone marrow, liver or kidney dysfunctions. Conversely, the attending physician would also know how to and when to adjust treatment to higher levels if the clinical response is not adequate (precluding toxic side effects) . Again, any suitable route of administration may be employed for providing the patient with an effective dosage of an arsenic compound. For example, oral, transdermal, iontophoretic, parenteral (subcutaneous, intramuscular, intrathecal and the like) may be employed. Dosage forms include tablets, troches, cachet, dispersions, suspensions, solutions, capsules, patches, and the like. (See,
Remington's Pharmaceutical Sciences.)
The pharmaceutical compositions of the present invention comprise an arsenic compound as the active ingredient, pharmaceutically acceptable salt thereof, and may also contain a pharmaceutically acceptable carrier, and optionally, other therapeutic ingredients, for example all trans retinoic acid. The term "pharmaceutically acceptable salts" refers to salts prepared from pharmaceutically acceptable non-toxic acids and bases, including inorganic and organic acids and bases.
The pharmaceutical compositions include compositions suitable for oral, mucosal routes, transdermal, iontophoretic, parenteral (including subcutaneous, intramuscular, intrathecal and intravenous) , although the most suitable route in any given case will depend on the nature and severity of the condition being treated. In the case where an intravenous injection or infusion composition is employed, a suitable dosage range for use is, e.g.. from about one to about 40 mg arsenic trioxide total daily; about 0.001 to about 10 mg arsenic trioxide per kg body weight total daily, or about 0.1 to about 10 mg melarsoprol per kg body weight total daily.
In addition, the arsenic carrier could be delivered via charged and uncharged matrices used as drug delivery devices such as cellulose acetate membranes, also through targeted delivery systems such as fusogenic liposomes attached to antibodies or specific antigens.
In practical use, an arsenic compound can be combined as the active ingredient in intimate admixture with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques. The carrier may take a wide variety of forms depending on the form of preparation desired for administration, e.g. , oral or parenteral (including tablets, capsules, powders, intravenous injections or infusions) . In preparing the compositions for oral dosage form any of the usual pharmaceutical media may be employed, e.g. , water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents, and the like; in the case of oral liquid preparations, e.g. , suspensions, solutions, elixirs, liposomes and aerosols; starches, sugars, micro- crystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents, and the like in the case of oral solid preparations e.g. , powders, capsules, and tablets. In preparing the compositions for parenteral dosage form, such as intravenous injection or infusion, similar pharmaceutical media may be employed, e.g. , water, glycols, oils, buffers, sugar, preservatives and the like know to those skilled in the art. Examples of such parenteral compositions include, but are not limited to Dextrose 5%w/v, normal saline or other solutions. The total dose of the arsenic compound may be administered in a vial of intravenous fluid, e.g. , ranging from about 2 ml to about 2000 ml. The volume of dilution fluid will vary according to the total dose administered. For example, arsenic trioxide supplied as a 10 ml aqueous solution at 1 mg/ml concentration is diluted in 10 to 500 ml of 5% dextrose solution, and used for intravenous infusion over a period of time ranging from about ten minutes to about four hours.
An exemplary course of treatment of a patient with leukemia, lymphoma, or solid cancer can involve daily administration by intravenous infusion of arsenic trioxide in an aqueous solution at a daily dose of about 0.01 to 1 mg arsenic trioxide per kg of body weight of the patient. Preferably, about 0.15 mg arsenic trioxide per kg body weight per day is used. The course of treatment may continue until bone marrow remission is observed or when side effects are becoming serious. The course of treatment may be repeated for up to ten times over approximately 10 months with a break of about 3 to 6 weeks in between courses . The post-remission course of treatment involves infusion of arsenic trioxide at a daily dose of about 0.15 mg per kg of body weight of the patient on a daily or weekdays-only basis for a cumulative total of 25 days.
5. EXAMPLES
Described below are examples of uses of the arsenic compounds of the invention in treatment of various types of leukemia. Through these and other experiments the arsenic trioxide formulation of the invention were found to be well- tolerated in humans. For example, three APL patients were given 10 mg of the arsenic trioxide formulation of the invention once daily (flat dose) intravenous dose. 5.1. ARSENIC TRIOXIDE AND MELARSOPROL INDUCE
APOPTOSIS IN MYELOID LEUKEMIA CELL LINES
The activity of As203 and melarsoprol against myeloid leukemia cell lines, including the APL cell line NB4- 306 (a retinoic acid-resistant cell line derived from NB4 that no longer expresses the intact PML-RARα fusion protein) , HL60, KG-1, and the myelomonocytic cell line U937 was investigated. To examine the role of PML in mediating arsenical activity, the inventors also tested these agents using murine embryonic fibroblasts (MEFs) and bone marrow (BM) progenitors in which the PML gene had been inactivated by homologous recombination. Unexpectedly, it is found that both compounds inhibited cell growth and induced apoptosis in all cell lines tested. Melarsoprol was more potent than As203 at equimolar concentrations ranging from 10"' to 10"= mol/L. As:03 relocalized PML and PML-RARα onto nuclear bodies, which was followed by PML degradation in NB4 as well as in HL60 and U937 cell lines. Although melarsoprol was more potent in inhibiting growth and inducing apoptosis, it did not affect PML and/or PML-RARα nuclear localization. Moreover, both As:03 and melarsoprol comparably inhibited growth and induced apoptosis of PML+/+ and PML-/-MEF, and inhibited colony- forming unit erythroid (CFU-E) and CFU granulocyte-monocyte formation in BM cultures of PML+/+ and PML-/- progenitors. A detailed description of the methods, materials, and results of these experiments is provided in Wang et al., Blood, 1998, 92:1497-1504.
Results from the experiments show that the cytotoxic effect of both arsenicals in these cell lines is not mediated by mechanisms that are dependent on PML or PML- RARα expression. In most lines, melarsoprol was somewhat more potent compared with As20:. in inhibiting growth and inducing apoptosis, and the effects of both drugs were dose dependent. As previously reported, it is confirmed that As203 relocalized PML protein onto nuclear bodies and induced PML and PML-RARα degradation in NB4 cells while triggering spoptosis. However, similar effects were also observed in HL60 and U937 cells which do not harbor the PML-RARα fusion gene. Moreover, melarsoprol induced apoptosis in all the cell lines tested without altering PML and/or PML-RARα.
The differentiating action of As03 and melarsoprol, appeared negligible in vitro, and did not appear to depend on the expression and/or modulation of PML and/or PML-RARα either. In fact, the small effect observed by the inventors in long-term cultures (up to 2 weeks) , was comparable in all the cell lines tested with both compounds.
It is also found that bcl-2 downregulation, which has been previously linked to the antileukemic effects of
As203 in APL, was also not dependent on expression of PML-RARα protein, because it occurred in the NB4 subclone 306 in which the intact protein is not detectable. Finally, to test whether PML expression was essential to the antileukemic effects of arsenicals, both agents were tested in mouse embryonic fibroblasts and BM cells from animals wherein wild- type PML had been eliminated by homologous recombination. In these cells wholly lacking PML expression, both As20? and melarsoprol were equally effective in inhibiting growth and inducing apoptosis, and both had similar effects on normal
CFU-E and CFU-GM colony formation. Moreover, no differences between wild-type and PML-/- cells were observed. Without being limited by any theory, together, these data strongly support theory that the antileukemic effects of these arsenicals occurs independently of both PML and PML-RARα expression. These results are in keeping with the medicinal history of arsenicals for diseases tnat are not characterized by alterations in PML protein such as, for instance, chronic myelocytic leukemia. The results indicate that both As:0> and melarsoprol are broadly active as antileukemic agents in both myeloid and lymphoid diseases. In conclusion, the data indicate that cytotoxic activity is not mediated by the PML protein and therefore is not limited to diseases that are associated with alterations in PML expression. Thus, the arsenic compounds of the invention have a potentially broader therapeutic role that is not confined to APL. 5.2. CLINICAL STUDY OF MELARSOPROL IN PATIENTS WITH ADVANCED LEUKEMIA
Melarsoprol, an organic arsenical synthesized by complexing elarsen oxide with dimercaprol , has primarily been used for the treatment of African trypanosomiasis. The effects of melarsoprol upon induction of apoptosis in cell lines representative of chronic B-cell lymphoproliferative disorders have been investigated, and the results are described below.
Melarsoprol (supplied as Arsobal [36 mg/mL] by Rhone Poulenc Rorer, Collegeville, PA) was diluted in propylene glycol at a stock concentration of 10"" mol/L and stored at room temperature. As20 (Sigma, St. Louis, MO) was dissolved in 1.65 mol/L sodium hydroxide (NaOH) at a stock solution of 10"- mol/L. Serial dilution (10"c to 10"' mol/L) were made in RPMI 1640 media. An Epstein-Barr virus (EBV)- transformed B-prolymphocytic cell line (JVM-2) , an EBV- transformed B-cell chronic lymphocytic leukemia (B-CLL) cell line (I83CLL) , and one non-EBV-transformed B-CLL cell line (WSU-CLL) were used as targets. Dose-response experiments with melarsoprol (10" to 10"' mol/L) were performed over 96 hours .
Unexpectedly, the inventors found that melarsoprol caused a dose- and time-dependent inhibition of survival and growth in all three cell lines. In contrast, As^O at similar concentrations had no effect on either viability or growth. After 24 hours, all three cell lines treated with melarsoprol (10" mol/L) exhibited morphologic characteristics of apoptosis. A prominent concentration-dependent downregulation of bcl-2 RNA after 24 hours of exposure to melarsoprol in WSU-CLL 183CLL, and -TVM-2 cells was observed. Decrease of bcl-2 protein expression was also observed in all three cell lines, whereas As20, had no effect on this parameter.
Given that the m vitro data above have shown unexpectedly broad antileukemic activity for melarsoprol against both myeloid and lymphoid cells, and generally at lower concentrations than As203 , a study was initiated to evaluate the pharmacokinetics, safety, and potential efficacy of melarsoprol in human patients with relapsed or refractory leukemia. Eligible patients were treated with a brief IV injection daily for 3 days, repeated weekly for 3 weeks, with an additional 3 wk course in responding pts. The initial dose was 1 mg/kg on Day 1, 2 mg/kg on Day 2, and 3.6 mg/kg on Day 3 and all days thereafter. Parallel in vitro studies included culture sensitivity of fresh leukemic cells to both melarsoprol and As^ , along with serial flow cytometric studies of surface antigen expression, apoptosis, and bcl-2 expression. Three patients with AML and one with CML have entered the study. Using a method based on high performance liquid chromatography that is sensitive to approximately 10 mg/ml, preliminary pharmacokinetic data show that peak plasma drug concentrations were obtained immediately after injection with a Cmax that ranged from 1.2 ng/ml on day 1 to 2.4 ng/ml on day 3. While the initial distribution phase was rapid, a prolonged T%γ has suggested release from a deep compartment. Plasma areas under the concentration x time curves (AUCs) were proportional to the administered dose, ranging from 0.48 ng-hr/ml on Day 1 to 1.48 ng-hr/ml on Day 3. Detectable concentrations of the drug were found in plasma one week after initial dosing. The drug has been relatively well- tolerated. Adverse effects have included transient pain at the injection site and mild nausea. No signs of "reactive encephalopathy" (occasionally observed during treatment of CNS trypanosomiasis) have been observed.
Results from these studies suggest that melarsoprol may have broader activity than inorganic As20, , and that concentrations which are cytotoxic to leukemic cells in vitro, and thus therapeutic, are readily achieved in vivo.
5.3. ARSENIC TRIOXIDE INDUCES APOPTOSIS IN K562 CHRONIC MYELOGENOUS LEUKEMIA (CML) CELLS A Philadelphia chromosome positive CML cell line K562 is used to determine if arsenic trioxide (As203) promotes apoptosis in CML. Suspension cultures of cells in log phase were exposed to As203 at concentrations of 1 x 10" M, 5 x 10_tlM, and 1 x 10"c M. Aliquots of cells were analyzed at various time points over the course of 72 hours to assess viability and apoptosis. Viability was measured using trypan blue exclusion; at the same time, apoptosis was detected by morphology, flow cytometry, and DNA gel electrophoresis.
Arsenic trioxide at a concentration of 1 x 10"D M had no effect on K562 cell growth or viability. The greatest effect on cell growth and survival was seen with 1 x 10"" M As203. K562 cell growth and viability data after 72 hours of exposure to As20 are recorded in Table 1 : Table 1:
% Cel l Growth Impairment % Viabil ity p value
Control 0 92 . 1 ± 0 . 9
5 x 10 '6 M As203 63 . 0 78 . 8 ± 0 . 5 0 . 0001 1 x 10"5 M As,θ3 75 . 3 61 . 9 ± 2 . 9 0 . 0223
Evidence that this arsenic-induced decrease in viability represented apoptosis was analyzed. Morphologic features of apoptosis including membrane blebbing and nuclear condensation were evident in stained cytospins of K562 cells incubated with 10"; M As20 for 72 hours. This correlated with evidence of DNA internucleosomal damage as visualized by gel electrophoresis of DNA extracted from K562 cells exposed to 10"" M As203. Quantitative assessment of apoptosis, as measured by the TUNEL method demonstrated that 75.6% ± 8.6 (1 x 10"3 M As203) cells exhibited apoptosis as compared with 6.3% ± 3.0 (control) cells at 72 hours. Treatment of K562 cells with 10"" M As20, resulted in an upregulation of p21 mRNA, as detected by Northern analysis, suggesting an arrest of the cells in the Gl phase of the cell cycle. This data indicates arsenic trioxide as a therapeutic agent for CML. 5.4. THERAPEUTIC TRIALS WITH RETINOIC ACID AND ARSENIC TRIOXIDE (As203) IN PML-RARα AND PLZF-RARα TRANSGENIC MICE Acute promyelocytic leukemia (APL) is associated with chromosomal translocations which invariably involve the translocation of the Retinoic Acid Receptor α (RARα) locus on chromosome 17 to other loci in the genome, such as in the majority of APL cases, the PML gene located on chromosome 15, and in a few cases the PLZF gene on chromosome 11. Patients harboring the t(15;17) are sensitive to treatment with All- Trans Retinoic Acid (ATRA) , yielding complete remission rates of 75% to 95%. APL associated with the t(ll;17) (PLZF-RARα) shows a poor response to ATRA. To test the efficacy of As20:. in the treatment of
APL, models of the disease were created in transgenic mice. Transgenic mice were generated by standard techniques in which the expression of the PML-RARα or PLZF-RARα fusion proteins is placed under the control of a myeloid- promyelocytic specific human Cathepsin-G (hCG) minigene.
Both hCG-PML/RARα and hCG-PLZF-RARα transgenic mice develop myeloid leukemia with features of APL similar to those in humans .
Therapeutic trials on these leukemic mice with the following regimens were started: 1) ATRA: 1.5 μg per gram of body weight per day administered orally; and 2) ATRA: 6 μg per gram of body weight per day administered intraperitoneally. Mice were bled once a week to evaluate the response. PML/RARα leukemias responded well to ATRA with high remission rates (80% with regimen 1) . Surprisingly, in vitro , ATRA induced differentiation, and inhibited growth of leukemic cells as well as leukemic colony formation in bone marrow and spleen progenitors assays in both PML-RARα and PLZF-RARα leukemias. Furthermore, in ex vivo experiments, leukemic cells from PLZF-RARα mice lost their tumorigenic capacity when transplanted in recipient nude mice upon pre- incubation with ATRA, while untreated cells were tumorigenic. However, in vivo , PLZF-RARα leukemias responded poorly to
ATRA (28% with regimen 1) , while higher doses of ATRA appeared more effective (50% with regimen 2) . In conclusion, leukemias in PLZF-RARα transgenic mice are sensitive to ATRA treatment, but might require therapeutic regimens with high doses of ATRA. These findings have direct implications in the treatment of APL patients with t(ll;17).
In both PML-RARα and PLZF-RARα leukemias, ATRA prolonged survival, but leukemias relapsed shortly after remission was achieved, and were refractory to further ATRA treatment. The two transgenic mouse models is also used to test the efficacy and dosage of As-0 , and ATRA+As:0, in combination for treatment of APL patients resistant to ATRA, and in APL associated with the t(ll;17). A regimen of As20- at 6 μg per day or a combination of As20: at 6 μg and ATRA at 1.5 or 6 μg per gram of body weight per day is administered intraperitoneally . Mice are bled weekly to evaluate the remission of the APL.
5.5. MANUFACTURE AND STABILITY OF PHARMACEUTICAL FORMULATION
Solid ultrapure arsenic trioxide (As20;) was solubilized in a solution of 5 M sodium hydroxide (NaOH) . The suspension was stirred at ambient temperature for 5 minutes which yielded a clear, homogenous solution. The As20 solution (2 mL, 1.0 M) was added to 393.6 mL of H:0 in a 500 ml Erlenmeyer flask, which yielded an As20 concentration of 1 mg/mL at pH = 12. A 5.0 M HCl solution was prepared by dilution of HCl (49.26 mL, 37% wt/wt, 10/15 M) with H20 (50.74 mL) in a 250 mL Erlenmeyer flask. The HCl solution was later transferred via syringe to a 1000 mL empty evacuated container. The As203 solution was back titrated with HCl (0.725 L, 5.0 M) to pH 8.0. Approximately 10 mL of the backtitrated As20, solution was filtered through a Millex-GS 0.22 μm filter unit and was added to each of approximately 30 sterile evacuated sterile vials. To make the pharmaceutical composition which would be injected intravenously into patients, 10 mL of this solution was withdrawn from two of the vials and was added to a 500 mL 5%-dextrose solution which yielded a final pH of 6.5.
The high purity of the bulk starting material was confirmed (see Table 1) by atomic absorptio etry. Duplicate samples of four intermediate or final-step solutions were assayed for total arsenic content. Assay bulk powder confirmed the extremely high purity of the starting material. Data for arsenic content of the intermediate and finished product solutions are presented in Table 2 below.
The data below show that the solutions are stable in that there does not appear to be any indication of weight loss of arsenic over time.
Table 2 Arsenic content (ppm) of intermediate formulation and finished product solution of arsenic trioxide.
Figure imgf000030_0001
* Identity of sample codes:
A-01: Intermediate product solution after initial solubilization in NaOH. A-02: Intermediate product solution prior to HCl titration. A-03: Intermediate product prior to Millex filtration. A-04: Finished product from sterile 10 ml fill vial immediately after manufacturing. A-05: Finished product from capped vials two months after manufacturing.
EXAMPLES : CLINICAL TRIALS IN APL PATIENTS
Arsenic trioxide was evaluated in patients with APL to determine whether this agent induced either cytodifferentiation or apoptosis. Twelve patients who had relapsed from extensive prior therapy were treated with arsenic trioxide at doses ranging from 0.06 to 0.2 mg/kg per day until a bone marrow remission was achieved. Bone marrow mononuclear cells were serially monitored by flow cytometry for immunophenotype, fluorescence in situ hybridization (FISH) , reverse transcription polymerase chain reaction (RT- PCR) assay for PML/RAR-α expression, and Western blot expression of the apoptosis-associated proteins, caspases 1, 2 and 3. The results showed that low-doses of arsenic trioxide are highly effective for inducing complete remission in relapsed patients with APL. Clinical response is associated with incomplete cytodifferentiation and induction of apoptosis with caspase activation in leukemic cells.
6.1. METHODS
Clinical protocol: Eligibility requirements included a diagnosis of APL confirmed by cytogenetics or fluorescence in situ hybridization (FISH) analysis for a t(15;17) translocation, or by reverse transcriptase polymerase reaction (RT-PCR) assay for PML/RAR-α. Patients must have relapsed from standard therapy that had included all-trans retinoic acid plus a combination of cytotoxic drugs. Signed informed consent was required, and the protocol was reviewed and approved by this center's institutional review board
Arsenic trioxide treatment: Arsenic trioxide was supplied as an aqueous solution in 10 ml vials containing 1 mg/ml of drug. The drug was further diluted in 500 ml of 5%-dextrose solution and infused intravenously over 2 to 4 hours once per day. While the initial cohort of patients received either 10 or 15 g/day as a flat dose, the referral of two children prompted the invention of a weight-based regimen (0.15 mg/kg/day) that was heretofore unknown. The drug was given daily until bone marrow remission was observed. Patients who achieved complete remission were eligible for treatment with additional courses of therapy 3 to 6 weeks after the preceding course. Subsequent courses were generally given at a dose of 0.15 mg/kg/day for a cumulative total of 25 days, administered either daily or on a weekdays-only schedule, for a maximum total of 6 courses over approximately 10 months. Monitoring during study: Patients with coagulopathy were transfused with platelets and fresh-frozen plasma to maintain the platelet count and fibrinogen at target levels > 50,000 cells/cu mm and > 100 mg/dL, respectively. Blood counts, coagulation studies, serum chemistry profiles, urinalyses, and electrocardiograms were serially obtained. A bone marrow aspiration and/or biopsy was performed at baseline and periodically thereafter until remission was documented. Conventional response criteria were observed, which included recovery of bone marrow to <. 5% blasts, peripheral blood leukocytes > 3,000 cells/cu mm, and platelets > 100,000 cells/cu mm.
Cellular immunophenotype studies: Heparinized bone marrow or blood samples were collected and mononuclear cells were isolated by Ficoll-Hypaque centrifugation. Surface membrane antigens were detected by direct immunofluorescence staining using fluorescein isethiocynate (FITC) or phycoerythrin conjugated monoclonal antibodies: CD16 (Leu 11a), CDllb, CD33 (Leu M9) , HLA-DR, CD45, and CD14 , purchased from either
Becton-Dickinson (Mountainview, CA) or Immunotech Immunology (Marseille, France) . Dual-color staining was performed by incubating cells simultaneously with two monoclonal antibodies, including CD33-PE/CDllb-FITC and CD33-PE/CD16- FITC. Negative controls using irrelevant monoclonal immunoglobulins of the same isotype were analyzed concurrently. Flow cytometric analyses were performed on an EPICS Profile II flow cytometer (Coulter Electronics) equipped with a 488 nm argon laser. Forward and side-scatter cell parameters were measured and combined with CD45/CD14 staining to identify populations of interest and to exclude monocytes from the analysis gate. The Multiparameter Data Acquisition and Display System (MDADS, Coulter Electronics) was used to acquire and analyze data.
Fluorescence in situ hybridization (FISH) : Selected specimens that had undergone immunofluorescence staining for CD33 and CDllb were sorted for cells that coexpressed both antigens using a FACStar Plus cell-sorter (Becton-Dickinson) . Separated cells were incubated in culture media at 37° C for one hour, treated with hypotonic solution 0.075M KC1 for 5 minutes, fixed in 3 : 1 methanol: acetic acid fixative, and air- dried. Interphase FISH was performed using a specific
PML/RAR-α translocation dual-color probe (Vysis; Downer's Grove, IL) . Briefly, DNA from interphase cells was denatured by immersing slides in a solution of 50% formamide/2xSSC at 73° C for 5 minutes; the slides were then dehydrated in alcohol and air dried. A mixture of probe in hybridization mixture was applied, covered with a cover slip, and sealed with rubber cement. Hybridization was carried out at 37° C in a moist chamber for approximately 12 to 16 hours. Following hybridization, unbound probe was removed by washing the slides at 45° C in 50% formamide/2xSSC solution three times for 10 minutes each, followed by a wash in 2xSSC/0.1 NP-40 solution at 45° C for 5 minutes. Slides were then air dried and counter-stained with 4 ' , 6-diamidino-2-phenylindole and covered with a glass coverslip. Analysis of interphase cells for fluorescent signals was performed with a
Photometries Sensys camera fitted to a Zeiss axioscope. A minimum of 300 cells was studied for each sample.
Western blot analysis: Cells were lysed in a buffer containing 50 mM Tris-HCl, 0.5 mM ethylene glycol [bis]- [aminioacyl] tetra acetic acid, 170 mM NaCl, ImM dithiothreitol, 0.2% NP-40, 0.01 U/mL aprotinin, 10 μg/mL leupeptin, lOμg/mL pepstatin, and 1 μM phenylmethylsulfonyl fluoride (all from Sigma) . The lysates were then sonicated using a ultrasonic homogenizer (471C series, Cole Parmer
Instruments, Chicago, IL) and centrifuged at 7,500 g (Sorvall Instruments, Newtown, CT) . Protein content of the lysates was determined using a BioRad Protein Assay Kit (Bio-Rad Laboratories, Hercules, CA) at 595 nm with a BSA standard. A sample buffer containing 10 % glycerol, 0.4 % SDS, 0.3 % bromphenol blue, 0.2 % pyronin Y, in lx stacking buffer (Tris base 0.5 M, 0.8 % SDS) , 20 % 2-mercaptoethanol, was added to the cell lysates, which were heat-denatured at 95°C for 3 min. Subsequently, 15 μg/lane of protein was loaded on a SDS- polyacrylamide gel containing 12.5% polyacrylamide and was size-fractionated by electrophoresis. Proteins were electroblotted onto Tras-Blot° transfer medium (Bio-Rad) and stained with Ponceau-S as an internal loading control.
Rabbit anti-human monoclonal antibodies, including caspase 1, caspase 2 (both from Santa Cruz Biotechnology, Santa Cruz, CA) , and caspase 3 (PharMingen, San Diego, CA) were added, and bound antibodies were detected using the ECL™ chemiluminescence detection system (Amersha , Arlington Heights, IL) . Protein bands were quantified by computer densitometry.
RT-PCR analysis for PML/RAR-α: RT-PCR was performed using methods previously described (Miller et al., 1992, Proc. Natl. Acad. Sci. 89:2694-8; Miller et al., 1993, Blood, 82:1689-94) .
6.2. RESULTS Patients: Twelve patients with relapsed or refractory APL were treated. All patients had received extensive prior therapy with retinoids and cytotoxic drugs (Table 3). Two patients had relapsed from allogeneic bone marrow transplantation, one of whom had also failed donor T-cell reinfusion. One patient was being maintained on hemodialysis for chronic renal failure.
Clinical Efficacy: Eleven of the 12 patients achieved a complete remission after arsenic trioxide treatment. The patient who entered on hemodialysis sustained an intracranial hemorrhage on day 1 and died on day 5. The median duration of therapy in responding patients was 33 days (range, 12 to 39 days), the median daily dose was 0.16 mg/kg (range, 0.06 to 0.2 mg/kg), and the median cumulative dose during induction was 360 mg (range, 160 to 515 mg) (Table 3) . Complete remission by all criteria was attained at a median time of 47 days (range, 24 to 83 days) after initiation of therapy. Remission by bone marrow criteria - the determining factor for discontinuing therapy - was achieved first, usually followed in sequence by recovery of peripheral blood leukocytes and platelets. Over the range of doses used in this study, no differences in efficacy or time to response were obvious. After 2 courses of therapy, 8 of 11 patients tested had converted their RT-PCR assays for PML/RAR-α from positive. to negative.
All 11 patients in complete remission completed at least 1 post-remission treatment course with arsenic trioxide. Four, two, and one patient each have completed a total of three, four and five treatment courses, respectively. The median duration of remission is 5+ months (range, 1 to 9+ months) . However, 3 of the 11 patients relapsed during the second treatment course; none of these patients had converted their RT-PCR assays, and each appeared to have rapidly acquired drug resistance. Two of these individuals have since expired from progressive leukemia.
Adverse Events: The clinical condition of patients in this study was highly variable, which reflected their extensive prior therapy. The protocol did not require hospitalization; three patients completed induction therapy entirely as outpatients, and one other individual was hospitalized solely for placement of a venous catheter. However, 8 patients were hospitalized for complications of leukemia, 5 of whom required transfer to an intensive care unit, endotracheal intubation, and assisted ventilation for complications that included pulmonary hemorrhage, renal failure, sepsis, graft vs. host disease, non-specific pulmonary infiltrates, or hypotension. One patient required insertion of a permanent pacemaker after second-degree heart block developed in the setting of severe metabolic acidosis, hyperkalemia, hypotension, and renal insufficiency. However, the heart block reversed despite rechallenge with further arsenic trioxide therapy. The drug was temporarily withheld due to serious intercurrent medical complications in 5 patients for a median of 2 days (range, 1 to 5 days) . Two patients developed symptoms similar to that of the "retinoic acid syndrome"; both were presumptively treated with dexamethasone and improved. Only 2 patients required no platelet transfusions whatsoever; the median number of platelet units transfused was 61 (range, 0 to 586 units) .
The median total peripheral blood leukocyte count at entry was 4,700 cells/cu mm (range, 500 to 144,000 cells/cu mm). Six patients developed leukocytosis (i.e., > 20,000 cells/cu mm) that ranged from 20,800 to 144,200 cells/cu mm. No additional therapy was administered to these patients, and the leukocytosis resolved in all cases without further intervention.
Common adverse reactions included lightheadedness during the infusion, fatigue, musculoskeletal pain, and mild hyperglycemia. Three patients developed dysesthias presumably due to peripheral neuropathy. However, 2 of these patients had been immobilized for prolonged periods during assisted ventilation, and the other patient had an antecedent neuropathic history.
Immunophenotype studies: APL is characterized by cells that express CD33, an antigen typically associated with primitive myeloid cells. Arsenic trioxide therapy induced a progressive decrease in the proportion of cells that solely expressed CD33, along with an increase in the proportion of cells that expressed CDllb, an antigen associated with mature myeloid elements. While these changes would be anticipated from any agent that induced remission in APL, arsenic trioxide also induced expression of cells that simultaneously expressed both antigens. In most cases, these dual- expressing cells dominated the myeloid cell population, and they persisted for extended periods after complete remission was achieved by clinical criteria.
Fluorescence in situ hybridization analysis: Bone marrow mononuclear cells taken from a patient both early and later in complete remission were sorted by flow cytometry for coexpression of CD33 and CDllb. Using fluorescence in situ hybridization (FISH) analysis, three hundred cells were examined early in remission. Similar to control APL cells, the majority of these cells yielded a hybrid signal, indicating a translocation between PML and RAR-α genes and their origin from the neoplastic clone. However, when cells from the same patient were again sorted using these same parameters later in remission, only the normal pattern of fluorescence signals was detected, indicating their derivation from normal hematopoietic progenitors.
Western blot analysis: Protein extracts from bone marrow mononuclear cells were serially examined by Western blot analysis. The analysis showed that the precursor forms of caspase 2 and caspase 3 were upregulated in vivo in response to arsenic trioxide treatment. Moreover, this treatment also induced expression of cleaved fragments of caspase 1, indicating activation of the enzyme. There is also some indication that expression of the cleaved form of caspase 3 is increased. The antibody used in these experiments does not react with the cleaved form of caspase 2.
6.3. DISCUSSION
In this study, with few exceptions, patients admitted to the trial had sustained multiple relapses and were resistant to conventional chemotherapy, retinoids, or bone marrow transplantation. At entry, patients in this study suffered from numerous leukemia-related complications, including respiratory failure, disseminated Varicella zoster infection, cavitary aspergillosis, chronic renal failure, and graft-vs. -host disease. Moreover, 5 of the 12 patients required admission to an intensive care unit for assisted ventilation and supportive care, but these complications were not directly related to arsenic trioxide therapy.
Virtually all patients with a confirmed diagnosis of APL attained remission without the early mortality associated with retinoid therapy. Although less commonly observed compared with all-trans retinoic acid treatment, arsenic trioxide induced striking leukocytosis in several patients. Upon withholding other cytotoxic drugs, the leukocytosis disappeared as patients attained remission. Despite 3 early relapses, 8 of 11 patients tested converted RT-PCR assays for PML/RAR-α (a molecular marker of residual disease) to negative, a phenomenon that is unusual after all- trans retinoic acid treatment alone. Finally, arsenic trioxide is active in APL over at least a three-fold dose range from 0.06 to 0.20 mg/kg.
All-trans retinoic acid induces "terminal" differentiation of APL cells, but the cytodifferentiating effects of arsenic trioxide appear to be incomplete. Arsenic induces a population of cells that simultaneously express surface antigens characteristic of both mature and immature cells (i.e. CDllb and CD33, respectively). Early during induction, these cells retain the t(15;17) translocation that characterizes APL. Unexpectedly, these cells persisted in the bone marrow despite the achievement of a clinically complete remission; however, later in remission, the coexpressing cells - while still readily detectable - were no longer positive by in situ hybridization. The morphologic appearance of leukemic cells during therapy is also far less distinctive than that observed during therapy with all-trans retinoic acid. In fact, leukemic cells from many patients αisplayed few morphologic changes for 10 or more days, after which the proportion of leukemic cells progressively decreased. Following "non-terminal" differentiation, arsenic trioxide appeared to induce apoptosis, coincident with increased expression and conversion of cysteine proteases (termed caspases) from inactive precursors to activated enzymes. The caspase pathway has only recently been characterized as an important pathway of programmed cell death. Initially recognized due to homology between the C. elegans protein ced-3 and mammalian interleukin-lβ converting enzyme (ICE) , the family of caspases now encompasses at least 10 different proteins that cleave a number of polypeptides. in leukemic cell lines, caspase activation is inducible with a number of cytotoxic agents, including all-trans retinoic acid. Since these enzymes induce widespread proteolysis, it is conceivable that PML/RAR-α is a caspase substrate. A final similarity shared by arsenic trioxide and all-trans retinoic acid is the rapid development of clinical resistance in some individuals. Leukemic cells taken from two patients who relapsed retained in vitro sensitivity over concentrations ranging from 10""M to 10" M. Relative arsenic resistance due to decreased intracellular transport has been described in association with down-regulation of membrane transporters encoded by the ars operon in bacterial cells. Resistance in mammalian cells is less well-characterized, but alterations in membrane transport or efflux are probably important factors.
In summary, arsenic trioxide induces complete remission in patients with APL who have relapsed from extensive prior therapy. This drug causes partial but incomplete cytodifferentiation of leukemic cells, followed by caspase activation and induction of apoptosis.
Table 3: Clinical characteristics and induction therapy results of patients with acute
Figure imgf000040_0002
Figure imgf000040_0001
All patients had previously received one or more courses of all-trans retinoic acid, plus an anthracycline antibiotic plus cytosine arabinoside. * Denotes individuals with proven retinoid resistance (i.e. lack of response during reinduction or relapse while on retinoid maintenance) ; t Denotes patient who died early. Other treatment: " mitoxantrone/etoposide; allogeneic bone marrow transplantation; : methotrexate/vincristine/6-mercaptopurine; 3 9-cis retinoic acid plus M195 (anti-CD33 monoclonal antibody) .
7. EXAMPLES: CLINICAL USE IN LYMPHOMA
Based upon the initial discovery of the antitumor effects of arsenic trioxide in vitro against B-cell lymphocyte lines, the inventors treated one patient with intermediate-grade large cell lymphoma who had relapsed from multiple forms of conventional therapy, including autologous bone marrow transplantation. Despite rapid progression of his disease prior to starting the arsenic trioxide therapy, treatment with arsenic trioxide effected a major (>50%) shrinkage in the size of his cancerous lymph nodes and spleen, which was also associated with a major improvement of his quality of life.
8. EXAMPLES: CLINICAL USE IN NON-HEMATOPOIETIC CANCER Arsenic trioxide was also used to treat colon cancer. In a preliminary test, one patient with colon cancer who received one treatment with arsenic trioxide shewed a major reduction in his serum CEA (carcinoembryonic antigen) level. The patient received daily intravenous infusion of 0.1-5 mg arsenic trioxide per kg body weigh per day for five days. A change in the level of CEA from 19,901 ng/ml to 15,266 ng/ml, a 23% reduction, was observed. It is well known that the a reduced level of serum CEA is associated with antitumor response. Clinical data confirms that arsenic trixoide can also be used to treat other non-hematopoietic cancer, such as colon cancer. 9. EXAMPLES; PHARMACOKINETICS STUDIES
Several dose-ranging studies were conducted to examine the pharmacokinetics (PK) and biological effects of As203 in patients with APL and in patients with other hematologic diseases. In patients with APL, marrow mononuclear cells were serially monitored by flow cytometry for immunophenotype, fluorescence in situ hybridization (FISH) , and Western blot expression of the apoptosis- associated proteins, caspases 1, 2 and 3. Cells that coexpressed CDllb and CD33, and which by FISH analysis carried the t(15;17) translocation, progressively increased during treatment and persisted early in complete remission. As203 also induced in vivo expression of the proenzymes of caspase 2 and caspase 3 , and activation of both caspase 1 and caspase 3. PK analysis of blood and urine for elemental arsenic (As) content showed that As was distributed in both plasma and red blood cell fractions of whole blood. Parallel elimination curves suggested that these 2 compartments were freely exchangeable, and decayed from peak values with initial half lives of about 60 mins. The mean AUC on day 1 was about 400 ng-hr/ml. Approximately 20% of the administered dose was recovered in urine within the first 24 hrs.
We then initiated a dose-ranging study in patients with diseases other than APL using a daily intravenous dosing schedule for a cumulative total of 25 days per treatment course every 3-5 weeks at dose levels of 0.1 and 0.15 mg per kg body weight per day. To date, 10 patients have been accrued, including patients with CLL (2 patients) , AML (3 patients) , lymphoma (4 patients) , and CML (1 patient) . Five patients were removed from the study early due to rapid progression, and 5 completed the planned 25-day course. Over this dose range, the drug has proved well-tolerated; adverse effects have included skin rash, lightheadedness during the infusion, fatigue, and QTc prolongation on EKG. Results from this ongoing study show that clinical use of As:03 induces partial differentiation and apoptosis in APL, but that the therapeutic effects of this agent are not confined to this disorder. The present invention is not to be limited in scope by the specific embodiments described herein. Indeed, various modifications of the invention in addition to those described herein will become apparent to those skilled in the art from the foregoing description. Such modifications are intended to fall within the scope of the appended claims.
Various publications are cited herein, the disclosures of which are incorporated by reference in their entireties.

Claims

WHAT IS CLAIMED IS:
1. A method for treating acute myelogenous leukemia in a human comprising administering to a human with acute myelogenous leukemia a therapeutically effective amount of arsenic trioxide.
2. A method for treating chronic myelogenous leukemia in a human comprising administering to a human with chronic myelogenous leukemia a therapeutically effective amount of arsenic trioxide.
3. A method for treating solid cancer in a human comprising administering to a human with a solid cancer a therapeutically effective amount of arsenic trioxide.
4. The method of claim 3 in which the solid cancer is cancer of the digestive tract, soft tissues, oesophagus, liver, stomach, colon, lung, skin, brain, bone, breast, or prostate gland.
5. A method for treating leukemia resistant to treatment with retinoids in a human comprising administering to a human in need thereof a therapeutically effective amount of arsenic trioxide or melarsoprol.
6. A method for treating leukemia, lymphoma, or solid tumors in a human, comprising administering to a human in need thereof a therapeutically effective amount of melarsoprol .
7. The method of claim 1 , 2 , 3 , 4 , or 5 , wherein about 2.5 to 4.5 mg of arsenic trioxide is administered per day.
8. The method of claim 1, 2, 3, 4, or 5 , wherein about 0.15 mg of arsenic trioxide per kg body weight of the human is administered per day.
9. The method of claim 1, 2, 3, 4, or 5, wherein arsenic trioxide is administered by intravenous infusion.
10. The method of claim 1, 2, or 5, wherein administration of arsenic trioxide is repeated daily until bone marrow remission is observed in the human.
11. The method of claim 10 further comprising repeating once to ten times the steps of suspending treatment for 3 to 6 weeks, and resuming daily administration of arsenic trioxide five to seven times a week for a cumulative total of 25 days.
12. The method of claim 1, 2, or 5 , wherein all- trans retinoic acid is also administered to the human.
13. The method of claim 3 or 4 wherein administration of arsenic trioxide is repeated daily for 5 days.
14. The method of claim 13 which is repeated once a month.
15. The method of claim 6, wherein about 0.5 to 5 mg of melarsoprol per kg body weight of the human is administered per day.
16. A method for manufacturing a sterile pharmaceutical composition suitable for administration to humans which comprises arsenic trioxide, said method comprising:
(a) solubilizing arsenic trioxide in an aqueous solution at pH greater than 12 ;
(b) neutralizing the arsenic trioxide solution with hydrochloric acid to pH about 8.5;
(c) diluting the arsenic trioxide solution from step (b) in a pharmaceutical carrier that stabilizes and lowers the pH to about 7; and (d) sterilizing the pharmaceutical composition.
17. A sterile pharmaceutical composition suitable for injection into humans which comprises arsenic trioxide and dextrose in a pharmaceutically acceptable carrier.
18. The pharmaceutical composition of claim 17 comprising 1 mg/ml of arsenic trioxide.
PCT/US1998/024024 1997-11-10 1998-11-10 Process for producing arsenic trioxide formulations and methods for treating cancer using arsenic trioxide or melarsoprol WO1999024029A1 (en)

Priority Applications (11)

Application Number Priority Date Filing Date Title
CA2309652A CA2309652C (en) 1997-11-10 1998-11-10 Process for producing arsenic trioxide formulations and methods for treating cancer using arsenic trioxide or melarsoprol
AU13973/99A AU747474C (en) 1997-11-10 1998-11-10 Process for producing arsenic trioxide formulations and methods for treating cancer using arsenic trioxide or melarsoprol
KR1020007005045A KR20010031938A (en) 1997-11-10 1998-11-10 Process for producing arsenic trioxide formulations and methods for treating cancer using arsenic trioxide or melarsoprol
JP2000520121A JP2001522799A (en) 1997-11-10 1998-11-10 Method for producing arsenic trioxide preparation and method for treating cancer using arsenic trioxide or melarsoprol
PL98343868A PL343868A1 (en) 1997-11-10 1998-11-10 Process for producing arsenic trioxide formulations and methods for treating cancer using arsenic trioxide or melarsoprol
NZ504585A NZ504585A (en) 1997-11-10 1998-11-10 Use of arsenic trioxide (and melarsoprol) formulations for treating solid cancer such as acute myeloblastic leukemia, acute monoblastic leukemia, acute erythroleukemic leukemia, acute megakaryosblastic leukemia, acute myelomonocytic leukemia or acute undifferentiated leukemia
EP98957803A EP1037625A4 (en) 1997-11-10 1998-11-10 Process for producing arsenic trioxide formulations and methods for treating cancer using arsenic trioxide or melarsoprol
BR9814857-5A BR9814857A (en) 1997-11-10 1998-11-10 Processes for treating leukemia, solid cancer, leukemia, lymphoma or solid tumors in a human, and for making a sterile pharmaceutical composition, and the respective sterile pharmaceutical composition
IL13605198A IL136051A0 (en) 1997-11-10 1998-11-10 Process for producing arsenic trioxide formulations and methods for treating cancer using arsenic trioxide or melarsoprol
NO20002409A NO20002409L (en) 1997-11-10 2000-05-09 Procedures for producing arsenic trioxide formulations and methods for treating cancer using arsenic trioxide or melarsoprol
AU2002300339A AU2002300339B2 (en) 1997-11-10 2002-07-31 Process For Producing Arsenic Trioxide Formulations And Methods For Treating Cancer Using Arsenic Trioxide Or Melarsoprol

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US6465597P 1997-11-10 1997-11-10
US60/064,655 1997-11-10

Publications (1)

Publication Number Publication Date
WO1999024029A1 true WO1999024029A1 (en) 1999-05-20

Family

ID=22057410

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1998/024024 WO1999024029A1 (en) 1997-11-10 1998-11-10 Process for producing arsenic trioxide formulations and methods for treating cancer using arsenic trioxide or melarsoprol

Country Status (20)

Country Link
US (22) US6770304B2 (en)
EP (3) EP2255800B1 (en)
JP (2) JP2001522799A (en)
KR (1) KR20010031938A (en)
CN (1) CN1285743A (en)
AU (1) AU747474C (en)
BR (1) BR9814857A (en)
CA (1) CA2309652C (en)
CY (1) CY1113856T1 (en)
DK (1) DK2255800T3 (en)
ES (2) ES2399480T3 (en)
HK (1) HK1150762A1 (en)
ID (1) ID25622A (en)
IL (1) IL136051A0 (en)
NO (1) NO20002409L (en)
NZ (1) NZ504585A (en)
PL (1) PL343868A1 (en)
PT (2) PT1964557E (en)
TR (1) TR200002243T2 (en)
WO (1) WO1999024029A1 (en)

Cited By (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0955052A1 (en) * 1998-05-08 1999-11-10 Ill-Ju Bae Arsenic hexaoxide (As4O6) for use in cancer therapy and its pharmaceutical composition
EP1166789A2 (en) * 2000-06-29 2002-01-02 Bae, Ill-Ju Arsenolite containing angiogenesis inhibitor
WO2003057012A2 (en) 2002-01-07 2003-07-17 Board Of Regents, The University Of Texas System S-dimethylarsino-thiosuccinic acid s-dimethylarsino-2-thiobenzoic acid s-(dimethylarsino) glutathione as treatments for cancer
EP1356820A1 (en) * 2002-04-26 2003-10-29 Institut National De La Sante Et De La Recherche Medicale (Inserm) DNA vaccine combined with an inducer of tumor cell apoptosis
WO2004032822A2 (en) 2002-10-09 2004-04-22 The University Of Hong Kong Formulation of oral compositions comprising arsenic trioxide and methods of use thereof
KR100456831B1 (en) * 2001-04-28 2004-11-10 주식회사 코미팜 Anticancer drug composition containing salt of meta-arsenite
EP1500398A1 (en) * 2003-07-21 2005-01-26 Sahajanand Biotech Private Limited Herbo-mineral formulation for refractory leukemias and lymphomas
FR2860719A1 (en) * 2003-10-10 2005-04-15 Assist Publ Hopitaux De Paris USE OF ORGANOSENED OR ORGANOSTIBIC DERIVATIVES FOR ANTICANCER PROPERTIES
SG112006A1 (en) * 2003-04-23 2005-06-29 Tty Biopharm Co Ltd Radioactive arsenic-containing compounds and their uses in the treatment of tumors
EP1729747A2 (en) * 2003-10-03 2006-12-13 Duke University Thiol reactive agents as a therapeutic modality
WO2007082104A2 (en) 2006-01-13 2007-07-19 The Texas A & M University System Compounds and methods for the treatment of cancer
US7405314B2 (en) 2004-07-16 2008-07-29 The Texas A&M University System Compounds and methods for the treatment of cancer
US7968738B2 (en) 2005-07-29 2011-06-28 Ziopharm Oncology, Inc. Compounds and methods for the treatment of cancer
US8252773B2 (en) 2007-11-02 2012-08-28 Ziopharm Oncology, Inc. Combination therapy with organic arsenicals
US8273379B2 (en) 1997-11-10 2012-09-25 Memorial Sloan-Kettering Cancer Center Process for producing arsenic trioxide formulations and methods for treating cancer using arsenic trioxide or melarsoprol
US8394422B2 (en) 2002-04-26 2013-03-12 Centre National De La Recherche Scientifique (Cnrs) Arsenic therapy for autoimmune and/or inflammatory diseases in mice and humans
US8497300B2 (en) 2002-04-26 2013-07-30 Centre National De La Recherche Scientifique (Cnrs) Arsenic therapy for APLS-type autoimmune lymph-oproliferative syndrome in mice and humans
WO2014045083A1 (en) 2012-09-20 2014-03-27 Bendale Yogesh Narayan Process for bio synthesis of nano arsenic trioxide and its use in treatment of diseases including cancers
US8796329B2 (en) 2006-09-29 2014-08-05 Ziopharm Oncology, Inc. Method for controlling angiogenesis in animals
US8906422B2 (en) 2002-10-09 2014-12-09 The University Of Hong Kong Method for inhibiting cancer using arsenic trioxide
WO2015101618A1 (en) * 2013-12-30 2015-07-09 INSERM (Institut National de la Santé et de la Recherche Médicale) Combination of an arsenic compound and at least one retinoid for treating acute myeloid leukemia
EP3388111A1 (en) 2008-08-20 2018-10-17 Solasia Pharma K.K. Organoarsenic compound for the treatment of cancer
EP3552611A4 (en) * 2016-12-09 2020-06-24 Chemas Co., Ltd. Pharmaceutical composition for preventing or treating cancer, comprising tetraarsenic hexoxide crystalline polymorph

Families Citing this family (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1044559C (en) * 1995-08-23 1999-08-11 哈尔滨医科大学附属第一医院 "Ailing" anticancer injection
IL135620A0 (en) 1997-10-15 2001-05-20 Polarx Biopharmaceuticals Inc Compositions and methods for the treatment of primary and metastic neoplastic diseases using arsenic compounds
CN1233476A (en) * 1998-04-24 1999-11-03 陆道培 Medicine for treating acute leukemia, and method for preparing same
ATE444309T1 (en) * 2000-08-08 2009-10-15 Immunomedics Inc IMMUNOTHERAPY FOR CRONIC MYELOCYTIC LEUKEMIA WITH NAKED ANTI-NCA-90 ANTIBODIES
CN1332672C (en) * 2001-03-15 2007-08-22 张鹏 Arsenic trioxide powder for injection
KR20030058019A (en) * 2001-12-29 2003-07-07 한국원자력연구소 Radiosensitizer containing arsenic trioxide as the active ingredient
KR20030075017A (en) * 2002-03-15 2003-09-22 한국원자력연구소 Angiogenesis inhibitor comprising arsenic trioxide
US20080089949A1 (en) * 2006-10-13 2008-04-17 Yok-Lam Kwong Method for treating cancer using oral arsenic trioxide
US20080166425A1 (en) * 2002-10-09 2008-07-10 The University Of Hong Kong Method for inhibiting cancer using arsenic trioxide
CN1304053C (en) * 2003-08-13 2007-03-14 上海第二医科大学附属瑞金医院 Composition medicine for curing acute early-young granulocytic leukemia
KR100632250B1 (en) 2004-02-16 2006-10-11 정태호 Anticancer agent containing arsenic compound as an active ingredient
US20050196464A1 (en) * 2004-03-03 2005-09-08 Tty Biopharm Company Limited Method and pharmaceutical composition for treatment of skin neoplasm
EP1721615A1 (en) 2005-05-09 2006-11-15 Komipharm International Co., Ltd. Pharmaceutical compositions comprising sodium or potassium arsenite for the treatment of urogenital cancer and its metastasis
US20060292243A1 (en) * 2005-06-24 2006-12-28 Shao-Chi Hsin Arsenic compounds for the treatment of the arsenic-sensitive blast-cell related diseases
WO2007035755A2 (en) * 2005-09-19 2007-03-29 Duke University Methods of treating hematological malignancies
US20080233207A1 (en) * 2006-01-04 2008-09-25 Sheptovitsky Yelena G Injectable and Infusable Mercury Compositions and Methods for Treating Cancer
US20080089951A1 (en) * 2006-10-11 2008-04-17 The University Of Hong Kong Method for Inhibiting Cancer Using Arsenic Trioxide
KR101009893B1 (en) * 2007-04-06 2011-01-20 주식회사 엘지화학 Novel method for preparing high purity heteropoly acid
EP2231684A4 (en) * 2007-12-12 2011-09-07 Ziopharm Oncology Inc Compounds and methods for the treatment of cancer
US8650648B2 (en) 2008-03-26 2014-02-11 Sophos Limited Method and system for detecting restricted content associated with retrieved content
US20090246291A1 (en) * 2008-03-27 2009-10-01 Angelika Burger Method and compositions for treatment of cancer
CA2772411C (en) * 2009-09-10 2017-08-22 Kominox, Inc. Cancer stem cell-targeted and drug resistant cancer therapy
US8795738B2 (en) 2009-11-12 2014-08-05 Board Of Regents Of The University Of Texas System Use of arsenic for cancer therapy protection
US8834938B2 (en) 2011-05-18 2014-09-16 Board Of Regents Of The University Of Texas System Use of arsenic for cancer therapy protection
US9231234B2 (en) 2012-03-30 2016-01-05 Panasonic Intellectual Property Management Co., Ltd. Cylindrical battery
CA2884051A1 (en) * 2012-10-08 2014-04-17 Zensun (Shanghai) Science & Technology, Ltd. Compositions and methods for treating heart failure in diabetic patients
ES2954081T3 (en) 2015-01-29 2023-11-20 Eupharma Pty Ltd Compositions containing arsenic for use in treatment methods
CA2975406A1 (en) 2015-02-01 2016-08-04 Kumar KURUMADDALI High surface-area lyophilized compositions comprising arsenic for oral administration in patients
KR102015858B1 (en) * 2015-10-23 2019-08-30 손영태 Pharmaceutical composition for immunity enhancement containing detoxicated arsenic trioxide
KR102548836B1 (en) 2016-02-25 2023-07-03 삼성디스플레이 주식회사 Display apparatus
US9700580B1 (en) * 2016-06-14 2017-07-11 Marguerite Harning Method for cancer treatment
IL265986B (en) 2016-12-01 2022-09-01 Eupharma Pty Ltd Pharmaceutical composition comprising an alkali metal and/or an alkaline earth metal diarsenic (iii) tetraoxide and uses thereof
CN108498542A (en) * 2017-02-24 2018-09-07 四川兴科蓉药业有限责任公司 β-As4S4Purposes in preparing the drug for treating Malignancy
WO2019109095A1 (en) * 2017-12-01 2019-06-06 Beth Israel Deaconess Medical Center, Inc. Arsenic trioxide and retinoic acid compounds for treatment of idh2-associated disorders
GB201800736D0 (en) * 2018-01-17 2018-02-28 St Georges Hospital Medical School Combination therapy for treatment of leukemia
WO2020117868A1 (en) * 2018-12-03 2020-06-11 Beth Israel-Deaconess Medical Center, Inc. Arsenic compounds and retinoic acid compounds for treatment of idh-associated disorders
WO2020117867A1 (en) * 2018-12-03 2020-06-11 Beth Israel-Deaconess Medical Center, Inc. Darinaparsin and retinoic acid compounds for treatment of idh-associated disorders
CN110101713A (en) * 2019-05-16 2019-08-09 上海交通大学医学院附属第九人民医院 A kind of application of arsenic trioxide composition
CN112675197A (en) * 2021-01-22 2021-04-20 中山大学孙逸仙纪念医院 Pharmaceutical composition for driving tumor by Hedgehog signal pathway of children
US11583552B2 (en) 2021-02-17 2023-02-21 Manoj Maniar Pharmaceutical formulation of arsenic trioxide

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994002108A1 (en) * 1992-07-24 1994-02-03 The Johns Hopkins University Chemotherapy for cancer

Family Cites Families (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US132275A (en) 1872-10-15 Improvement in medical compounds
US232807A (en) 1880-10-05 Herbert e
US3700498A (en) 1970-12-10 1972-10-24 Ibm Process for making electrophotographic plates
JPS5188620A (en) 1975-01-31 1976-08-03 Jintainokenkonoiji kotonigannitaishitekonoojusuru noshukuionekinoseizoho
DE3003635C2 (en) 1980-02-01 1985-07-11 Klöckner-Humboldt-Deutz AG, 5000 Köln Process and device for dearsenic materials containing arsenic
US4501275A (en) * 1981-07-06 1985-02-26 Maahs Jerry D Mammalian subject heating unit using radiant heat
FR2539993A1 (en) 1983-01-28 1984-08-03 Darcheux Mario Balsam which acts on cancerous wounds
US4950768A (en) 1984-01-16 1990-08-21 Cronyn Marshall W Cyclic disulfonic ester cross-linking compounds
US4696814A (en) * 1985-08-21 1987-09-29 Warner-Lambert Company Parenteral phenytoin compositions
US4705030A (en) * 1987-02-17 1987-11-10 Tepperberg Phillip S Hand augmenting spinal manipulator encircling the hand
US4882142A (en) 1988-12-19 1989-11-21 The Dow Chemical Company Bone marrow suppressing agents
US5759837A (en) * 1989-01-17 1998-06-02 John Hopkins University Chemotherapy for cancer by inhibiting the fatty acid biosynthetic pathway
IL93618A0 (en) * 1989-03-06 1990-12-23 Lilly Co Eli Improved diluent formulation for daptomycin
CN1061908A (en) 1991-12-21 1992-06-17 江西省妇产医院 A kind of manufacture method of curing cancer drug
CN1060935C (en) 1992-05-31 2001-01-24 丛繁滋 Preparation method of arsenical capable of applying to cancer focus
CN1044777C (en) 1992-07-06 1999-08-25 杨世泽 Analgesic plaster for cancer pain and its preparing method
DE4224534A1 (en) 1992-07-24 1994-01-27 Marika Dr Med Ehrlich Anti-ovulation agent for hormonal contraception
DE4317331A1 (en) 1993-05-25 1994-12-01 Reischle Karl Georg Treatment of impairments of the immune system
ITTO930510A1 (en) 1993-07-09 1993-10-07 Walter Tarello ACTIVE DRUGS AGAINST CHRONIC FATIGUE SYNDROME (C.F.S.)
US5455270A (en) * 1993-08-11 1995-10-03 Bristol-Myers Squibb Co. Stabilized solutions of platinum(II) antitumor agents
CA2183547C (en) 1994-02-18 2002-05-14 Tamara Vasilievna Vorobieva Compositions for treating defective cell functions
US5646159A (en) 1994-07-20 1997-07-08 Research Triangle Institute Water-soluble esters of camptothecin compounds
CN1119113A (en) 1994-09-22 1996-03-27 衣永德 Compound Aifukang capsule for curing cancer and preparing process thereof
CN1122700A (en) 1994-11-08 1996-05-22 付知中 Medicinal powder for curing mastosis, tumor and skin and external diseases
LV11667B (en) 1995-08-14 1997-06-20 Tamara Vorobieva Remedy for renewal of disordered function of immunomodulation and cell tissue reproduction adjustors
CN1044559C (en) 1995-08-23 1999-08-11 哈尔滨医科大学附属第一医院 "Ailing" anticancer injection
DE69620802T2 (en) 1995-09-12 2002-10-10 Liposome Co Inc HYDROLYZABLE HYDROPHOBIC TAXAN DERIVATIVES
CN1058620C (en) 1995-11-12 2000-11-22 卢颖 Yingfengaidi medicine and its production process
CN1052648C (en) 1995-12-03 2000-05-24 唐书生 Repellent analgesic ointment for cancer or tumor
IL135620A0 (en) * 1997-10-15 2001-05-20 Polarx Biopharmaceuticals Inc Compositions and methods for the treatment of primary and metastic neoplastic diseases using arsenic compounds
AU2006202364B2 (en) 1997-11-10 2009-05-28 Sloan-Kettering Institute For Cancer Research Process for producing arsenic trioxide formulations and methods for treating cancer using arsenic trioxide or melarsoprol
EP2255800B1 (en) 1997-11-10 2012-10-17 Memorial Sloan Kettering Cancer Center Arsenic trioxide for use in the treatment of leukaemia
AU2002300339B2 (en) 1997-11-10 2006-03-02 Sloan-Kettering Institute For Cancer Research Process For Producing Arsenic Trioxide Formulations And Methods For Treating Cancer Using Arsenic Trioxide Or Melarsoprol
CN1233476A (en) 1998-04-24 1999-11-03 陆道培 Medicine for treating acute leukemia, and method for preparing same

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994002108A1 (en) * 1992-07-24 1994-02-03 The Johns Hopkins University Chemotherapy for cancer

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
CHEN G-Q, ET AL.: "IN VITRO STUDIES ON CELLULAR AND MOLECULAR MECHANISMS OF ARSENIC TROXIDE (AS2O3) IN THE TREATMENT OF ACUTE PROMYELOCYTIC LEUKEMIA: AS2O3 INDUCES NB4 CELL APOPTOSIS WITH DOWNREGULATION OF BEL-2 EXPRESSION AND MODULATION OF PML-RARALPHA/PML PROTEINS", BLOOD, AMERICAN SOCIETY OF HEMATOLOGY, US, vol. 88, no. 03, 1 August 1996 (1996-08-01), US, pages 1052 - 1061, XP002917127, ISSN: 0006-4971 *
See also references of EP1037625A4 *
SHIMOTSUURA S: "STUDIES ON THE ANTINEOPLASMIC ACTIONS OF AS2O3", SHIKA GAKUHO - JOURNAL OF TOKYO DENTAL COLLEGE SOCIETY, TOKYO SHIKA DAIGAKU GAKKAI,TOKYO,, JP, vol. 86, no. 08, 1 January 1986 (1986-01-01), JP, pages 1237 - 1253, XP002917125, ISSN: 0037-3710 *
ZHANG P, ET AL.: "TREATMENT OF ACUTE PROMYELOCYTIC LEUKEMIA WITH INTRAVENOUS ARSENIC TRIOXIDE", ZHONGHUA XUEYEXUE ZAZHI - CHINESE JOURNAL OF HEMATOLOGY, TIANJIN,, CN, vol. 17, no. 02, 1 February 1996 (1996-02-01), CN, pages 58 - 60, XP002917126, ISSN: 0253-2727 *

Cited By (50)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8273379B2 (en) 1997-11-10 2012-09-25 Memorial Sloan-Kettering Cancer Center Process for producing arsenic trioxide formulations and methods for treating cancer using arsenic trioxide or melarsoprol
EP0955052A1 (en) * 1998-05-08 1999-11-10 Ill-Ju Bae Arsenic hexaoxide (As4O6) for use in cancer therapy and its pharmaceutical composition
EP1166789A2 (en) * 2000-06-29 2002-01-02 Bae, Ill-Ju Arsenolite containing angiogenesis inhibitor
KR100399657B1 (en) * 2000-06-29 2003-09-29 배일주 Angiogenesis Inhibitor
EP1166789A3 (en) * 2000-06-29 2003-10-22 Bae, Ill-Ju Arsenolite containing angiogenesis inhibitor
US6703049B2 (en) 2000-06-29 2004-03-09 Il Ju Bae Angiogenesis inhibitor
KR100456831B1 (en) * 2001-04-28 2004-11-10 주식회사 코미팜 Anticancer drug composition containing salt of meta-arsenite
US7619000B2 (en) 2002-01-07 2009-11-17 The Texas A&M University System S-dimethylarsino-thiosuccinic acid S-dimethylarsino-2-thiobenzoic acid S-(dimethylarsino) glutathione as treatments for cancer
US6911471B2 (en) 2002-01-07 2005-06-28 The Texas A&M University System S-dimethylarsino-thiosuccinic acid s-dimethylarsino-2-thiobenzoic acid s-(dimethylarsino) glutathione as treatments for cancer
EP2420229A1 (en) 2002-01-07 2012-02-22 Board Of Regents, University Of Texas System S-dimethylarsino-thiosuccinic acid s-dimethylarsino-2-thiobenzoic acid s-(dimethylarsino) glutathione as treatments for cancer
WO2003057012A3 (en) * 2002-01-07 2004-07-08 Univ Texas S-dimethylarsino-thiosuccinic acid s-dimethylarsino-2-thiobenzoic acid s-(dimethylarsino) glutathione as treatments for cancer
EP2462932A1 (en) 2002-01-07 2012-06-13 Board Of Regents, The University Of Texas System S-dimethylarsino-thiosuccinic acid s-dimethylarsino-2-thiobenzoic acid s-(dimethylarsino) glutathione as treatments for cancer
WO2003057012A2 (en) 2002-01-07 2003-07-17 Board Of Regents, The University Of Texas System S-dimethylarsino-thiosuccinic acid s-dimethylarsino-2-thiobenzoic acid s-(dimethylarsino) glutathione as treatments for cancer
US6995188B2 (en) 2002-01-07 2006-02-07 Board Of Regents, The University Of Texas System S-dimethylarsino-thiosuccinic acid s-dimethylarsino-2-thiobenzoic acid s-(dimethylarsino) glutathione as treatments for cancer
US8394422B2 (en) 2002-04-26 2013-03-12 Centre National De La Recherche Scientifique (Cnrs) Arsenic therapy for autoimmune and/or inflammatory diseases in mice and humans
US8497300B2 (en) 2002-04-26 2013-07-30 Centre National De La Recherche Scientifique (Cnrs) Arsenic therapy for APLS-type autoimmune lymph-oproliferative syndrome in mice and humans
EP2272527A3 (en) * 2002-04-26 2011-07-06 INSERM (Institut National de la Santé et de la Recherche Médicale) Combined DNA vaccine and biological modifiers for cancer therapy
EP1356820A1 (en) * 2002-04-26 2003-10-29 Institut National De La Sante Et De La Recherche Medicale (Inserm) DNA vaccine combined with an inducer of tumor cell apoptosis
WO2003090778A2 (en) * 2002-04-26 2003-11-06 Institute National De La Sante Et De La Recherche Medicale (Inserm) Dna vaccine combined with an inducer of tumor cell apoptosis
WO2003090778A3 (en) * 2002-04-26 2004-01-22 Inst Nat Sante Rech Med Dna vaccine combined with an inducer of tumor cell apoptosis
EP3106169A1 (en) * 2002-10-09 2016-12-21 Versitech Limited Formulation of oral compositions comprising arsenic trioxide and methods of use thereof
WO2004032822A2 (en) 2002-10-09 2004-04-22 The University Of Hong Kong Formulation of oral compositions comprising arsenic trioxide and methods of use thereof
US8906422B2 (en) 2002-10-09 2014-12-09 The University Of Hong Kong Method for inhibiting cancer using arsenic trioxide
US7521071B2 (en) * 2002-10-09 2009-04-21 Versitech Limited Formulation of oral compositions comprising arsenic trioxide and methods of use thereof
EP1562616B1 (en) * 2002-10-09 2016-06-01 Versitech Limited Formulation of oral compositions comprising arsenic trioxide and methods of use thereof
EP1562616A2 (en) * 2002-10-09 2005-08-17 The University of Hong Kong Formulation of oral compositions comprising arsenic trioxide and methods of use thereof
US7803350B2 (en) 2003-04-23 2010-09-28 Institute Of Nuclear Energy Research Rocaec Radioactive arsenic-containing compounds and their uses in the treatment of tumors
SG112006A1 (en) * 2003-04-23 2005-06-29 Tty Biopharm Co Ltd Radioactive arsenic-containing compounds and their uses in the treatment of tumors
EP1500398A1 (en) * 2003-07-21 2005-01-26 Sahajanand Biotech Private Limited Herbo-mineral formulation for refractory leukemias and lymphomas
EP1729747A2 (en) * 2003-10-03 2006-12-13 Duke University Thiol reactive agents as a therapeutic modality
EP1729747A4 (en) * 2003-10-03 2008-12-10 Univ Duke Thiol reactive agents as a therapeutic modality
FR2860719A1 (en) * 2003-10-10 2005-04-15 Assist Publ Hopitaux De Paris USE OF ORGANOSENED OR ORGANOSTIBIC DERIVATIVES FOR ANTICANCER PROPERTIES
WO2005034935A1 (en) * 2003-10-10 2005-04-21 Assistance Publique - Hopitaux De Paris Use of organoarsenic or organoantimony derivatives for their anticancer properties
US7405314B2 (en) 2004-07-16 2008-07-29 The Texas A&M University System Compounds and methods for the treatment of cancer
US7968738B2 (en) 2005-07-29 2011-06-28 Ziopharm Oncology, Inc. Compounds and methods for the treatment of cancer
EP2394702A1 (en) 2005-07-29 2011-12-14 Ziopharm Oncology, Inc. Compounds and methods for the treatment of cancer
US8334398B2 (en) 2005-07-29 2012-12-18 Ziopharm Oncology, Inc. Compounds and methods for the treatment of cancer
US8865764B2 (en) 2005-07-29 2014-10-21 Solasia Pharma K.K. Compounds and methods for the treatment of cancer
WO2007082104A2 (en) 2006-01-13 2007-07-19 The Texas A & M University System Compounds and methods for the treatment of cancer
US8796329B2 (en) 2006-09-29 2014-08-05 Ziopharm Oncology, Inc. Method for controlling angiogenesis in animals
US8252773B2 (en) 2007-11-02 2012-08-28 Ziopharm Oncology, Inc. Combination therapy with organic arsenicals
US11324713B2 (en) 2008-08-20 2022-05-10 Solasia Pharma K.K. Organoarsenic compounds and methods for the treatment of cancer
EP3388111A1 (en) 2008-08-20 2018-10-17 Solasia Pharma K.K. Organoarsenic compound for the treatment of cancer
US11324714B2 (en) 2008-08-20 2022-05-10 Solasia Pharma K.K. Organoarsenic compounds and methods for the treatment of cancer
US10842769B2 (en) 2008-08-20 2020-11-24 Solasia Pharma K.K. Organoarsenic compounds and methods for the treatment of cancer
WO2014045083A1 (en) 2012-09-20 2014-03-27 Bendale Yogesh Narayan Process for bio synthesis of nano arsenic trioxide and its use in treatment of diseases including cancers
WO2015101618A1 (en) * 2013-12-30 2015-07-09 INSERM (Institut National de la Santé et de la Recherche Médicale) Combination of an arsenic compound and at least one retinoid for treating acute myeloid leukemia
US11077139B2 (en) 2013-12-30 2021-08-03 Inserm (Institut National De La Sante Et De La Recherche Medicale) Combination of an arsenic compound and at least one retinoid for treating acute myeloid leukemia
US11191779B2 (en) 2016-12-09 2021-12-07 Chemas Co., Ltd. Pharmaceutical composition for preventing or treating cancer, comprising tetraarsenic hexoxide crystalline polymorph
EP3552611A4 (en) * 2016-12-09 2020-06-24 Chemas Co., Ltd. Pharmaceutical composition for preventing or treating cancer, comprising tetraarsenic hexoxide crystalline polymorph

Also Published As

Publication number Publication date
US6982096B2 (en) 2006-01-03
EP1964557B1 (en) 2013-01-02
US20040146577A1 (en) 2004-07-29
NO20002409D0 (en) 2000-05-09
US20040146570A1 (en) 2004-07-29
US20110091573A1 (en) 2011-04-21
US20040146572A1 (en) 2004-07-29
US20040146575A1 (en) 2004-07-29
BR9814857A (en) 2001-12-26
US20040146573A1 (en) 2004-07-29
US20040146582A1 (en) 2004-07-29
IL136051A0 (en) 2001-05-20
HK1150762A1 (en) 2012-01-13
AU747474B2 (en) 2002-05-16
NO20002409L (en) 2000-06-27
US7879364B2 (en) 2011-02-01
CY1113856T1 (en) 2016-07-27
US6723351B2 (en) 2004-04-20
US20030211171A1 (en) 2003-11-13
AU747474C (en) 2006-09-21
PT1964557E (en) 2013-02-20
US6884439B2 (en) 2005-04-26
EP2255800B1 (en) 2012-10-17
KR20010031938A (en) 2001-04-16
EP1964557A1 (en) 2008-09-03
US20040146568A1 (en) 2004-07-29
PT2255800E (en) 2012-12-17
AU1397399A (en) 1999-05-31
US20040146583A1 (en) 2004-07-29
ID25622A (en) 2000-10-19
US6855339B2 (en) 2005-02-15
DK2255800T3 (en) 2013-02-04
PL343868A1 (en) 2001-09-10
US20020013371A1 (en) 2002-01-31
US20040146580A1 (en) 2004-07-29
US20040146574A1 (en) 2004-07-29
CN1285743A (en) 2001-02-28
TR200002243T2 (en) 2000-12-21
US6770304B2 (en) 2004-08-03
CA2309652C (en) 2013-01-29
US20060029681A1 (en) 2006-02-09
JP2010184926A (en) 2010-08-26
US6861076B2 (en) 2005-03-01
US8273379B2 (en) 2012-09-25
EP1037625A1 (en) 2000-09-27
JP2001522799A (en) 2001-11-20
NZ504585A (en) 2002-06-28
US20040146569A1 (en) 2004-07-29
US20040146571A1 (en) 2004-07-29
US20030099719A1 (en) 2003-05-29
EP2255800A1 (en) 2010-12-01
US20040146576A1 (en) 2004-07-29
US20040146578A1 (en) 2004-07-29
US20040146581A1 (en) 2004-07-29
US20040146579A1 (en) 2004-07-29
EP1037625A4 (en) 2002-07-31
ES2395515T3 (en) 2013-02-13
US20040151782A1 (en) 2004-08-05
CA2309652A1 (en) 1999-05-20
ES2399480T3 (en) 2013-04-01

Similar Documents

Publication Publication Date Title
AU747474C (en) Process for producing arsenic trioxide formulations and methods for treating cancer using arsenic trioxide or melarsoprol
AU2006202364B2 (en) Process for producing arsenic trioxide formulations and methods for treating cancer using arsenic trioxide or melarsoprol
AU2002300339B2 (en) Process For Producing Arsenic Trioxide Formulations And Methods For Treating Cancer Using Arsenic Trioxide Or Melarsoprol
MXPA00004460A (en) Process for producing arsenic trioxide formulations and methods for treating cancer using arsenic trioxide or melarsoprol

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 136051

Country of ref document: IL

Ref document number: 98813016.5

Country of ref document: CN

AK Designated states

Kind code of ref document: A1

Designated state(s): AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GD GE GH GM HR HU ID IL IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG UZ VN YU ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW SD SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
ENP Entry into the national phase

Ref document number: 2309652

Country of ref document: CA

Ref document number: 2309652

Country of ref document: CA

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: PA/a/2000/004460

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2000 520121

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 1020007005045

Country of ref document: KR

Ref document number: 13973/99

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 1998957803

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 504585

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 2000/02243

Country of ref document: TR

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: 1998957803

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1020007005045

Country of ref document: KR

WWG Wipo information: grant in national office

Ref document number: 13973/99

Country of ref document: AU

WWW Wipo information: withdrawn in national office

Ref document number: 1020007005045

Country of ref document: KR